101
|
Mhatre-Winters I, Eid A, Han Y, Tieu K, Richardson JR. Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Astrocytes from Humanized Targeted Replacement Mice. ASN Neuro 2023; 15:17590914221144549. [PMID: 36604975 PMCID: PMC9982390 DOI: 10.1177/17590914221144549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Apolipoprotein E4 (APOE4) genotype and sex are significant risk factors for Alzheimer's disease (AD), with females demonstrating increased risk modulated by APOE genotype. APOE is predominantly expressed in astrocytes, however, there is a lack of comprehensive assessments of sex differences in astrocytes stratified by APOE genotype. Here, we examined the response of mixed-sex and sex-specific neonatal APOE3 and APOE4 primary mouse astrocytes (PMA) to a cytokine mix of IL1b, TNFa, and IFNg. Pro-inflammatory and anti-inflammatory cytokine profiles were assessed by qRT-PCR and Meso Scale Discovery multiplex assay. Mixed-sex APOE4 PMA were found to have higher basal messenger RNA expression of several pro-inflammatory cytokines including Il6, Tnfa, Il1b, Mcp1, Mip1a, and Nos2 compared to APOE3 PMA, which was accompanied by increased levels of these secreted cytokines. In sex-specific cultures, basal expression of Il1b, Il6, and Nos2 was 1.5 to 2.5 fold higher in APOE4 female PMA compared to APOE4 males, with both being higher than APOE3 PMA. Similar results were found for secreted levels of these cytokines. Together, these findings indicate that APOE4 genotype and female sex, contribute to a greater inflammatory response in primary astrocytes and these data may provide a framework for investigating the mechanisms contributing to genotype and sex differences in AD-related neuroinflammation.
Collapse
Affiliation(s)
- Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA,Department of Neurosciences, School of Biomedical Sciences, Kent
State University, Kent, OH, USA
| | - Aseel Eid
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Yoonhee Han
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Jason R. Richardson
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA,Jason R. Richardson, Department of
Environmental Health Sciences, Robert Stempel College of Public Health and
Social Work, Florida International University, Miami, FL 33199-2156, USA.
| |
Collapse
|
102
|
Wartchow KM, Scaini G, Quevedo J. Glial-Neuronal Interaction in Synapses: A Possible Mechanism of the Pathophysiology of Bipolar Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:191-208. [PMID: 36949311 DOI: 10.1007/978-981-19-7376-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Bipolar disorder (BD) is a severe and chronic psychiatric disorder that affects approximately 1-4% of the world population and is characterized by recurrent episodes of mania or hypomania and depression. BD is also associated with illnesses marked by immune activation, such as metabolic syndrome, obesity, type 2 diabetes mellitus, and cardiovascular diseases. Indeed, a connection has been suggested between neuroinflammation and peripheral inflammatory markers in the pathophysiology of BD, which can be associated with the modulation of many dysfunctional processes, including synaptic plasticity, neurotransmission, neurogenesis, neuronal survival, apoptosis, and even cognitive/behavioral functioning. Rising evidence suggests that synaptic dysregulations, especially glutamatergic system dysfunction, are directly involved in mood disorders. It is becoming clear that dysregulations in connection and structural changes of glial cells play a central role in the BD pathophysiology. This book chapter highlighted the latest findings that support the theory of synaptic dysfunction in BD, providing an overview of the alterations in neurotransmitters release, astrocytic uptake, and receptor signaling, as well as the role of inflammation on glial cells in mood disorders. Particular emphasis is given to the alterations in presynaptic and postsynaptic neurons and glial cells, all cellular elements of the "tripartite synapse," compromising the neurotransmitters system, excitatory-inhibitory balance, and neurotrophic states of local networks in mood disorders. Together, these studies provide a foundation of knowledge about the exact role of the glial-neuronal interaction in mood disorders.
Collapse
Affiliation(s)
- Krista M Wartchow
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Giselli Scaini
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - João Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
103
|
Snapper DM, Reginauld B, Liaudanskaya V, Fitzpatrick V, Kim Y, Georgakoudi I, Kaplan DL, Symes AJ. Development of a novel bioengineered 3D brain-like tissue for studying primary blast-induced traumatic brain injury. J Neurosci Res 2023; 101:3-19. [PMID: 36200530 DOI: 10.1002/jnr.25123] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 11/08/2022]
Abstract
Primary blast injury is caused by the direct impact of an overpressurization wave on the body. Due to limitations of current models, we have developed a novel approach to study primary blast-induced traumatic brain injury. Specifically, we employ a bioengineered 3D brain-like human tissue culture system composed of collagen-infused silk protein donut-like hydrogels embedded with human IPSC-derived neurons, human astrocytes, and a human microglial cell line. We have utilized this system within an advanced blast simulator (ABS) to expose the 3D brain cultures to a blast wave that can be precisely controlled. These 3D cultures are enclosed in a 3D-printed surrogate skull-like material containing media which are then placed in a holder apparatus inside the ABS. This allows for exposure to the blast wave alone without any secondary injury occurring. We show that blast induces an increase in lactate dehydrogenase activity and glutamate release from the cultures, indicating cellular injury. Additionally, we observe a significant increase in axonal varicosities after blast. These varicosities can be stained with antibodies recognizing amyloid precursor protein. The presence of amyloid precursor protein deposits may indicate a blast-induced axonal transport deficit. After blast injury, we find a transient release of the known TBI biomarkers, UCHL1 and NF-H at 6 h and a delayed increase in S100B at 24 and 48 h. This in vitro model will enable us to gain a better understanding of clinically relevant pathological changes that occur following primary blast and can also be utilized for discovery and characterization of biomarkers.
Collapse
Affiliation(s)
- Dustin M Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, USA
| | - Bianca Reginauld
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, USA
| | - Volha Liaudanskaya
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Yeonho Kim
- Preclinical Behavior and Modeling Core, Uniformed Services University, Bethesda, Maryland, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, USA
| |
Collapse
|
104
|
The current state of carnivore cognition. Anim Cogn 2023; 26:37-58. [PMID: 36333496 DOI: 10.1007/s10071-022-01709-2] [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: 06/15/2022] [Revised: 10/10/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
The field of animal cognition has advanced rapidly in the last 25 years. Through careful and creative studies of animals in captivity and in the wild, we have gained critical insights into the evolution of intelligence, the cognitive capacities of a diverse array of taxa, and the importance of ecological and social environments, as well as individual variation, in the expression of cognitive abilities. The field of animal cognition, however, is still being influenced by some historical tendencies. For example, primates and birds are still the majority of study species in the field of animal cognition. Studies of diverse taxa improve the generalizability of our results, are critical for testing evolutionary hypotheses, and open new paths for understanding cognition in species with vastly different morphologies. In this paper, we review the current state of knowledge of cognition in mammalian carnivores. We discuss the advantages of studying cognition in Carnivorans and the immense progress that has been made across many cognitive domains in both lab and field studies of carnivores. We also discuss the current constraints that are associated with studying carnivores. Finally, we explore new directions for future research in studies of carnivore cognition.
Collapse
|
105
|
Barzilai A, Mitiagin Y. Ataxia-telangiectasia mutated plays an important role in cerebellar integrity and functionality. Neural Regen Res 2023; 18:497-502. [DOI: 10.4103/1673-5374.350194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
106
|
Bennison SA, Liu X, Toyo-Oka K. Nuak kinase signaling in development and disease of the central nervous system. Cell Signal 2022; 100:110472. [PMID: 36122883 DOI: 10.1016/j.cellsig.2022.110472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 01/14/2023]
Abstract
Protein kinases represent important signaling hubs for a variety of biological functions. Many kinases are traditionally studied for their roles in cancer cell biology, but recent advances in neuroscience research show repurposed kinase function to be important for nervous system development and function. Two members of the AMP-activated protein kinase (AMPK) related family, NUAK1 and NUAK2, have drawn attention in neuroscience due to their mutations in autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), schizophrenia, and intellectual disability (ID). Furthermore, Nuak kinases have also been implicated in tauopathy and other disorders of aging. This review highlights what is known about the Nuak kinases in nervous system development and disease and explores the possibility of Nuak kinases as targets for therapeutic innovation.
Collapse
Affiliation(s)
- Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Xiaonan Liu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
107
|
Phadke L, Lau DHW, Aghaizu ND, Ibarra S, Navarron CM, Granat L, Magno L, Whiting P, Jolly S. A primary rodent triculture model to investigate the role of glia-neuron crosstalk in regulation of neuronal activity. Front Aging Neurosci 2022; 14:1056067. [DOI: 10.3389/fnagi.2022.1056067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Neuroinflammation and hyperexcitability have been implicated in the pathogenesis of neurodegenerative disease, and new models are required to investigate the cellular crosstalk involved in these processes. We developed an approach to generate a quantitative and reproducible triculture system that is suitable for pharmacological studies. While primary rat cells were previously grown in a coculture medium formulated to support only neurons and astrocytes, we now optimised a protocol to generate tricultures containing neurons, astrocytes and microglia by culturing in a medium designed to support all three cell types and adding exogenous microglia to cocultures. Immunocytochemistry was used to confirm the intended cell types were present. The percentage of ramified microglia in the tricultures decreases as the number of microglia present increases. Multi-electrode array recordings indicate that microglia in the triculture model suppress neuronal activity in a dose-dependent manner. Neurons in both cocultures and tricultures are responsive to the potassium channel blocker 4-aminopyridine, suggesting that neurons remained viable and functional in the triculture model. Furthermore, suppressed neuronal activity in tricultures correlates with decreased densities of dendritic spines and of the postsynaptic protein Homer1 along dendrites, indicative of a direct or indirect effect of microglia on synapse function. We thus present a functional triculture model, which, due to its more complete cellular composition, is a more relevant model than standard cocultures. The model can be used to probe glia-neuron interactions and subsequently aid the development of assays for drug discovery, using neuronal excitability as a functional endpoint.
Collapse
|
108
|
Munger EL, Edler MK, Hopkins WD, Hof PR, Sherwood CC, Raghanti MA. Comparative analysis of astrocytes in the prefrontal cortex of primates: Insights into the evolution of human brain energetics. J Comp Neurol 2022; 530:3106-3125. [PMID: 35859531 PMCID: PMC9588662 DOI: 10.1002/cne.25387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
Astrocytes are the main homeostatic cell of the brain involved in many processes related to cognition, immune response, and energy expenditure. It has been suggested that the distribution of astrocytes is associated with brain size, and that they are specialized in humans. To evaluate these, we quantified astrocyte density, soma volume, and total glia density in layer I and white matter in Brodmann's area 9 of humans, chimpanzees, baboons, and macaques. We found that layer I astrocyte density, soma volume, and ratio of astrocytes to total glia cells were highest in humans and increased with brain size. Overall glia density in layer I and white matter were relatively invariant across brain sizes, potentially due to their important metabolic functions on a per volume basis. We also quantified two transporters involved in metabolism through the astrocyte-neuron lactate shuttle, excitatory amino acid transporter 2 (EAAT2) and glucose transporter 1 (GLUT1). We expected these transporters would be increased in human brains due to their high rate of metabolic consumption and associated gene activity. While humans have higher EAAT2 cell density, GLUT1 vessel volume, and GLUT1 area fraction compared to baboons and chimpanzees, they did not differ from macaques. Therefore, EAAT2 and GLUT1 are not related to increased energetic demands of the human brain. Taken together, these data provide evidence that astrocytes play a unique role in both brain expansion and evolution among primates, with an emphasis on layer I astrocytes having a potentially significant role in human-specific metabolic processing and cognition.
Collapse
Affiliation(s)
- Emily L. Munger
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, OH
| | - Melissa K. Edler
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, OH
| | - William D. Hopkins
- Department of Comparative Medicine, University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chet C. Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Mary Ann Raghanti
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, OH
| |
Collapse
|
109
|
Cunha F, Stingo-Hirmas D, Cardoso RF, Wright D, Henriksen R. Neuronal and non-neuronal scaling across brain regions within an intercross of domestic and wild chickens. Front Neuroanat 2022; 16:1048261. [PMID: 36506870 PMCID: PMC9732670 DOI: 10.3389/fnana.2022.1048261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/11/2022] [Indexed: 11/26/2022] Open
Abstract
The allometric scaling of the brain size and neuron number across species has been extensively studied in recent years. With the exception of primates, parrots, and songbirds, larger brains have more neurons but relatively lower neuronal densities than smaller brains. Conversely, when considering within-population variability, it has been shown that mice with larger brains do not necessarily have more neurons but rather more neurons in the brain reflect higher neuronal density. To what extent this intraspecific allometric scaling pattern of the brain applies to individuals from other species remains to be explored. Here, we investigate the allometric relationships among the sizes of the body, brain, telencephalon, cerebellum, and optic tectum, and the numbers of neurons and non-neuronal cells of the telencephalon, cerebellum, and optic tectum across 66 individuals originated from an intercross between wild and domestic chickens. Our intercross of chickens generates a population with high variation in brain size, making it an excellent model to determine the allometric scaling of the brain within population. Our results show that larger chickens have larger brains with moderately more neurons and non-neuronal cells. Yet, absolute number of neurons and non-neuronal cells correlated strongly and positively with the density of neurons and non-neuronal cells, respectively. As previously shown in mice, this scaling pattern is in stark contrast with what has been found across different species. Our findings suggest that neuronal scaling rules across species are not a simple extension of the neuronal scaling rules that apply within a species, with important implications for the evolutionary developmental origins of brain diversity.
Collapse
|
110
|
Wang L, Yu C, Tao Y, Yang X, Jiang Q, Yu H, Zhang J. Transcriptome analysis reveals potential marker genes for diagnosis of Alzheimer's disease and vascular dementia. Front Genet 2022; 13:1038585. [PMID: 36506318 PMCID: PMC9730885 DOI: 10.3389/fgene.2022.1038585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VD) are the two most common forms of dementia, share similar symptoms, and are sometimes difficult to distinguish. To investigate the potential mechanisms by which they differ, we identified differentially expressed genes in blood and brain samples from patients with these diseases, and performed weighted gene co-expression network analysis and other bioinformatics analyses. Weighted gene co-expression network analysis resulted in mining of different modules based on differences in gene expression between these two diseases. Enrichment analysis and generation of a protein-protein interaction network were used to identify core pathways for each disease. Modules were significantly involved in cAMP and AMPK signaling pathway, which may be regulated cell death in AD and VD. Genes of cAMP and neurotrophin signaling pathways, including ATP1A3, PP2A, NCEH1, ITPR1, CAMKK2, and HDAC1, were identified as key markers. Using the least absolute shrinkage and selection operator method, a diagnostic model for AD and VD was generated and verified through analysis of gene expression in blood of patients. Furthermore, single sample gene set enrichment analysis was used to characterize immune cell infiltration into brain tissue. That results showed that infiltration of DCs and pDCs cells was increased, and infiltration of B cells and TFH cells was decreased in the brain tissues of patients with AD and VD. In summary, classification based on target genes showed good diagnostic efficiency, and filled the gap in the diagnostic field or optimizes the existing diagnostic model, which could be used to distinguish between AD and VD.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, The Second Affiliated Hospital of the Harbin Medical University, Harbin, China
| | - Chunjiang Yu
- Department of Neurology, The Second Affiliated Hospital of the Harbin Medical University, Harbin, China
| | - Ye Tao
- Department of Neurology, The First Hospital of SuiHua City, Suihua, China
| | - Xiumei Yang
- Department of Cardiovascularology, The Fifth Hospital of the Harbin City, Harbin, China
| | - Qiao Jiang
- Department of Neurology, The Fifth People’s Hospital of the Dalian City, Dalian, China
| | - Haiyu Yu
- Rehabilitation Department of Jiamusi Center Hospital, Jiamusi, China
| | - Jiejun Zhang
- Department of Neurology, Hebei Yanda Hospital, Hebei, China
| |
Collapse
|
111
|
Borgenheimer E, Hamel K, Sheeler C, Moncada FL, Sbrocco K, Zhang Y, Cvetanovic M. Single nuclei RNA sequencing investigation of the Purkinje cell and glial changes in the cerebellum of transgenic Spinocerebellar ataxia type 1 mice. Front Cell Neurosci 2022; 16:998408. [PMID: 36457352 PMCID: PMC9706545 DOI: 10.3389/fncel.2022.998408] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Glial cells constitute half the population of the human brain and are essential for normal brain function. Most, if not all, brain diseases are characterized by reactive gliosis, a process by which glial cells respond and contribute to neuronal pathology. Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disease characterized by a severe degeneration of cerebellar Purkinje cells (PCs) and cerebellar gliosis. SCA1 is caused by an abnormal expansion of CAG repeats in the gene Ataxin1 (ATXN1). While several studies reported the effects of mutant ATXN1 in Purkinje cells, it remains unclear how cerebellar glia respond to dysfunctional Purkinje cells in SCA1. To address this question, we performed single nuclei RNA sequencing (snRNA seq) on cerebella of early stage Pcp2-ATXN1[82Q] mice, a transgenic SCA1 mouse model expressing mutant ATXN1 only in Purkinje cells. We found no changes in neuronal and glial proportions in the SCA1 cerebellum at this early disease stage compared to wild-type controls. Importantly, we observed profound non-cell autonomous and potentially neuroprotective reactive gene and pathway alterations in Bergmann glia, velate astrocytes, and oligodendrocytes in response to Purkinje cell dysfunction.
Collapse
Affiliation(s)
- Ella Borgenheimer
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Katherine Hamel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Carrie Sheeler
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | | - Kaelin Sbrocco
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ying Zhang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, United States
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
112
|
Hwang S, Lee Y, Jun SB. Co-culture platform for neuron-astrocyte interaction using optogenetic modulation. Biomed Eng Lett 2022; 12:401-411. [PMID: 36238374 PMCID: PMC9550905 DOI: 10.1007/s13534-022-00243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/01/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022] Open
Abstract
For decades, the role of glial cells has attracted attention in the neuroscience field. Particularly, although the astrocyte is the most abundant glial cell type, it was believed to function as a passive support cell. However, recent evidence suggests that astrocytes actively release various gliotransmitters and signaling entities that regulate the excitability of pre-and post-synaptic neurons in the brain. In this study, we optimized the ratio of astrocytes and neurons to investigate the interaction between astrocytes and neurons. To this end, postnatal day 0-1 rodent hippocampi were dissociated and cultured. The neuron-astrocyte ratio was monitored for up to 3 weeks after treating the cultures with 0, 1, and 5 µM of cytosine arabinoside (Ara-C) at DIV 2. Subsequently, from postnatal transgenic (TG) mouse expressing ChR2 on astrocytes, hippocampi were cultured on the microelectrode array (MEA) with the desired neuron-astrocyte ratio. The astrocyte was irradiated using a 473 nm blue laser for 30 s in a cycle of 10 Hz and electrophysiological recording was performed to verify the activities of neurons induced by the stimulated astrocytes. Astrocytes and neurons in both co-cultures increased at an identical ratio when treated with 1 µM Ara-C, whereas they decreased significantly when treated with 5 µM Ara-C. Particularly, the laser-stimulated astrocytes induced an increase in the frequency of neuronal activities and lasted after illumination. The proposed co-culture platform is expected to be used in experiments to investigate the network between astrocytes and neurons in vitro.
Collapse
Affiliation(s)
- Seoyoung Hwang
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Korea
| | - Yena Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Korea
| | - Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Korea
- Graduate Program in Smart Factory, Ewha Womans University, Seoul, 03760 Korea
- Department of Brain and Cognitive Science, College of Scranton, Ewha Womans University, Seoul, 03760 Korea
| |
Collapse
|
113
|
Yang L, Lu J, Guo J, Chen J, Xiong F, Wang X, Chen L, Yu C. Ventrolateral Periaqueductal Gray Astrocytes Regulate Nociceptive Sensation and Emotional Motivation in Diabetic Neuropathic Pain. J Neurosci 2022; 42:8184-8199. [PMID: 36109166 PMCID: PMC9636999 DOI: 10.1523/jneurosci.0920-22.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 11/21/2022] Open
Abstract
Diabetic neuropathic pain (DNP) is a diabetes complication experienced by many patients. Ventrolateral periaqueductal gray (vlPAG) neurons are essential mediators of the descending pain modulation system, yet the role of vlPAG astrocytes in DNP remains unclear. The present study applied a multidimensional approach to elucidate the role of these astrocytes in DNP. We verified the activation of astrocytes in different regions of the PAG in male DNP-model rats. We found that only astrocytes in the vlPAG exhibited increased growth. Furthermore, we described differences in vlPAG astrocyte activity at different time points during DNP progression. After the 14th day of modeling, vlPAG astrocytes exhibited obvious activation and morphologic changes. Furthermore, activation of Gq-designer receptors exclusively activated by a designer drug (Gq-DREADDs) in vlPAG astrocytes in naive male rats induced neuropathic pain-like symptoms and pain-related aversion, whereas activation of Gi-DREADDs in vlPAG astrocytes in male DNP-model rats alleviated sensations of pain and promoted pain-related preference behavior. Thus, bidirectional manipulation of vlPAG astrocytes revealed their potential to regulate pain. Surprisingly, activation of Gi-DREADDs in vlPAG astrocytes also mitigated anxiety-like behavior induced by DNP. Thus, our results provide direct support for the hypothesis that vlPAG astrocytes regulate diabetes-associated neuropathic pain and concomitant anxiety-like behavior.SIGNIFICANCE STATEMENT Many studies examined the association between the ventrolateral periaqueductal gray (vlPAG) and neuropathic pain. However, few studies have focused on the role of vlPAG astrocytes in diabetic neuropathic pain (DNP) and DNP-related emotional changes. This work confirmed the role of vlPAG astrocytes in DNP by applying a more direct and robust approach. We used chemogenetics to bidirectionally manipulate the activity of vlPAG astrocytes and revealed that vlPAG astrocytes regulate DNP and pain-related behavior. In addition, we discovered that activation of Gi-designer receptors exclusively activated by a designer drug in vlPAG astrocytes alleviated anxiety-like behavior induced by DNP. Together, these findings provide new insights into DNP and concomitant anxiety-like behavior and supply new therapeutic targets for treating DNP.
Collapse
Affiliation(s)
- Lan Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Jingshan Lu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Jianpeng Guo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Jian Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Fangfang Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Xinyao Wang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Li Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, 350122, Fujian China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, 350122, Fujian China
| |
Collapse
|
114
|
Li ZQ, Li TX, Tian M, Ren ZS, Yuan CY, Yang RK, Shi SJ, Li H, Kou ZZ. Glial cells and neurologic autoimmune disorders. Front Cell Neurosci 2022; 16:1028653. [PMID: 36385950 PMCID: PMC9644207 DOI: 10.3389/fncel.2022.1028653] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/03/2022] [Indexed: 12/01/2023] Open
Abstract
Neurologic autoimmune disorders affect people's physical and mental health seriously. Glial cells, as an important part of the nervous system, play a vital role in the occurrence of neurologic autoimmune disorders. Glial cells can be hyperactivated in the presence of autoantibodies or pathological changes, to influence neurologic autoimmune disorders. This review is mainly focused on the roles of glial cells in neurologic autoimmune disorders and the influence of autoantibodies produced by autoimmune disorders on glial cells. We speculate that the possibility of glial cells might be a novel way for the investigation and therapy of neurologic autoimmune disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hui Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an, China
| | - Zhen-Zhen Kou
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
115
|
Saito ER, Warren CE, Hanegan CM, Larsen JG, du Randt JD, Cannon M, Saito JY, Campbell RJ, Kemberling CM, Miller GS, Edwards JG, Bikman BT. A Novel Ketone-Supplemented Diet Improves Recognition Memory and Hippocampal Mitochondrial Efficiency in Healthy Adult Mice. Metabolites 2022; 12:1019. [PMID: 36355101 PMCID: PMC9693360 DOI: 10.3390/metabo12111019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 09/13/2023] Open
Abstract
Mitochondrial dysfunction and cognitive impairment are common symptoms in many neurologic and psychiatric disorders, as well as nonpathological aging. Ketones have been suggested as therapeutic for their efficacy in epilepsy and other brain pathologies such as Alzheimer's disease and major depressive disorder. However, their effects on cognitive function in healthy individuals is less established. Here, we explored the mitochondrial and performative outcomes of a novel eight-week ketone-supplemented ketogenic (KETO) diet in healthy adult male and female mice. In a novel object recognition test, KETO mice spent more time with the novel, compared to familiar, object, indicating an improvement in recognition memory. High-resolution respirometry on permeabilized hippocampal tissue returned significant reductions in mitochondrial O2 consumption. No changes in ATP production were observed, yielding a significantly higher ATP:O2 ratio, a measure of mitochondrial efficiency. Together, these findings demonstrate the KETO diet improves hippocampal mitochondrial efficiency. They add to a growing body of evidence that suggests ketones and ketogenic diets are neuroprotective and metabolically and cognitively relevant, even in healthy adults. They also suggest that ketogenic lifestyle changes may be effective strategies for protecting against cognitive decline associated with aging and disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Benjamin T. Bikman
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
116
|
Bullock DN, Hayday EA, Grier MD, Tang W, Pestilli F, Heilbronner SR. A taxonomy of the brain's white matter: twenty-one major tracts for the 21st century. Cereb Cortex 2022; 32:4524-4548. [PMID: 35169827 PMCID: PMC9574243 DOI: 10.1093/cercor/bhab500] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/26/2023] Open
Abstract
The functional and computational properties of brain areas are determined, in large part, by their connectivity profiles. Advances in neuroimaging and network neuroscience allow us to characterize the human brain noninvasively, but a comprehensive understanding of the human brain demands an account of the anatomy of brain connections. Long-range anatomical connections are instantiated by white matter, which itself is organized into tracts. These tracts are often disrupted by central nervous system disorders, and they can be targeted by neuromodulatory interventions, such as deep brain stimulation. Here, we characterized the connections, morphology, traversal, and functions of the major white matter tracts in the brain. There are major discrepancies across different accounts of white matter tract anatomy, hindering our attempts to accurately map the connectivity of the human brain. However, we are often able to clarify the source(s) of these discrepancies through careful consideration of both histological tract-tracing and diffusion-weighted tractography studies. In combination, the advantages and disadvantages of each method permit novel insights into brain connectivity. Ultimately, our synthesis provides an essential reference for neuroscientists and clinicians interested in brain connectivity and anatomy, allowing for the study of the association of white matter's properties with behavior, development, and disorders.
Collapse
Affiliation(s)
- Daniel N Bullock
- Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University Bloomington, Bloomington, IN 47405, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Elena A Hayday
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark D Grier
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Wei Tang
- Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University Bloomington, Bloomington, IN 47405, USA
- Department of Computer Science, Indiana University Bloomington, Bloomington, IN 47408, USA
| | - Franco Pestilli
- Department of Psychology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Sarah R Heilbronner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
117
|
Devan SP, Jiang X, Luo G, Xie J, Quirk JD, Engelbach JA, Harmsen H, McKinley ET, Cui J, Zu Z, Attia A, Garbow JR, Gore JC, McKnight CD, Kirschner AN, Xu J. Selective Cell Size MRI Differentiates Brain Tumors from Radiation Necrosis. Cancer Res 2022; 82:3603-3613. [PMID: 35877201 PMCID: PMC9532360 DOI: 10.1158/0008-5472.can-21-2929] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/05/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Brain metastasis is a common characteristic of late-stage lung cancers. High doses of targeted radiotherapy can control tumor growth in the brain but can also result in radiotherapy-induced necrosis. Current methods are limited for distinguishing whether new parenchymal lesions following radiotherapy are recurrent tumors or radiotherapy-induced necrosis, but the clinical management of these two classes of lesions differs significantly. Here, we developed, validated, and evaluated a new MRI technique termed selective size imaging using filters via diffusion times (SSIFT) to differentiate brain tumors from radiotherapy necrosis in the brain. This approach generates a signal filter that leverages diffusion time dependence to establish a cell size-weighted map. Computer simulations in silico, cultured cancer cells in vitro, and animals with brain tumors in vivo were used to comprehensively validate the specificity of SSIFT for detecting typical large cancer cells and the ability to differentiate brain tumors from radiotherapy necrosis. SSIFT was also implemented in patients with metastatic brain cancer and radiotherapy necrosis. SSIFT showed high correlation with mean cell sizes in the relevant range of less than 20 μm. The specificity of SSIFT for brain tumors and reduced contrast in other brain etiologies allowed SSIFT to differentiate brain tumors from peritumoral edema and radiotherapy necrosis. In conclusion, this new, cell size-based MRI method provides a unique contrast to differentiate brain tumors from other pathologies in the brain. SIGNIFICANCE This work introduces and provides preclinical validation of a new diffusion MRI method that exploits intrinsic differences in cell sizes to distinguish brain tumors and radiotherapy necrosis.
Collapse
Affiliation(s)
- Sean P Devan
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, 37232, USA
| | - Xiaoyu Jiang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Guozhen Luo
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jingping Xie
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James D Quirk
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO 63110, USA
| | - John A Engelbach
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO 63110, USA
| | - Hannah Harmsen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Eliot T McKinley
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Jing Cui
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Albert Attia
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Joel R Garbow
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO 63110, USA
- Alvin J Siteman Cancer Center, Washington University, St. Louis, MO, 63110, USA
| | - John C. Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37232, USA
| | - Colin D McKnight
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Austin N Kirschner
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Junzhong Xu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
118
|
Zhang NN, Zhang Y, Wang ZZ, Chen NH. Connexin 43: insights into candidate pathological mechanisms of depression and its implications in antidepressant therapy. Acta Pharmacol Sin 2022; 43:2448-2461. [PMID: 35145238 PMCID: PMC9525669 DOI: 10.1038/s41401-022-00861-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/06/2022] [Indexed: 11/09/2022]
Abstract
Major depressive disorder (MDD), a chronic and recurrent disease characterized by anhedonia, pessimism or even suicidal thought, remains a major chronic mental concern worldwide. Connexin 43 (Cx43) is the most abundant connexin expressed in astrocytes and forms the gap junction channels (GJCs) between astrocytes, the most abundant and functional glial cells in the brain. Astrocytes regulate neurons' synaptic strength and function by expressing receptors and regulating various neurotransmitters. Astrocyte dysfunction causes synaptic abnormalities, which are related to various mood disorders, e.g., depression. Increasing evidence suggests a crucial role of Cx43 in the pathogenesis of depression. Depression down-regulates Cx43 expression in humans and rats, and dysfunction of Cx43 also induces depressive behaviors in rats and mice. Recently Cx43 has received considerable critical attention and is highly implicated in the onset of depression. However, the pathological mechanisms of depression-like behavior associated with Cx43 still remain ambiguous. In this review we summarize the recent progress regarding the underlying mechanisms of Cx43 in the etiology of depression-like behaviors including gliotransmission, metabolic disorders, and neuroinflammation. We also discuss the effects of antidepressants (monoamine antidepressants and ketamine) on Cx43. The clarity of the candidate pathological mechanisms of depression-like behaviors associated with Cx43 and its potential pharmacological roles for antidepressants will benefit the exploration of a novel antidepressant target.
Collapse
Affiliation(s)
- Ning-Ning Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
119
|
Hawkinson TR, Clarke HA, Young LEA, Conroy LR, Markussen KH, Kerch KM, Johnson LA, Nelson PT, Wang C, Allison DB, Gentry MS, Sun RC. In situ spatial glycomic imaging of mouse and human Alzheimer's disease brains. Alzheimers Dement 2022; 18:1721-1735. [PMID: 34908231 PMCID: PMC9198106 DOI: 10.1002/alz.12523] [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: 06/06/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 01/28/2023]
Abstract
N-linked protein glycosylation in the brain is an understudied facet of glucose utilization that impacts a myriad of cellular processes including resting membrane potential, axon firing, and synaptic vesicle trafficking. Currently, a spatial map of N-linked glycans within the normal and Alzheimer's disease (AD) human brain does not exist. A comprehensive analysis of the spatial N-linked glycome would improve our understanding of brain energy metabolism, linking metabolism to signaling events perturbed during AD progression, and could illuminate new therapeutic strategies. Herein we report an optimized in situ workflow for enzyme-assisted, matrix-assisted laser desorption and ionization (MALDI) mass spectrometry imaging (MSI) of brain N-linked glycans. Using this workflow, we spatially interrogated N-linked glycan heterogeneity in both mouse and human AD brains and their respective age-matched controls. We identified robust regional-specific N-linked glycan changes associated with AD in mice and humans. These data suggest that N-linked glycan dysregulation could be an underpinning of AD pathologies.
Collapse
Affiliation(s)
- Tara R. Hawkinson
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Harrison A. Clarke
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Lyndsay E. A. Young
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Lindsey R. Conroy
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Kia H. Markussen
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Kayla M. Kerch
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Lance A. Johnson
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Peter T. Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Derek B. Allison
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Matthew S. Gentry
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Ramon C. Sun
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
120
|
The Reactive Astrocytes After Surgical Brain Injury Potentiates the Migration, Invasion, and Angiogenesis of C6 Glioma. World Neurosurg 2022; 168:e595-e606. [DOI: 10.1016/j.wneu.2022.10.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
|
121
|
Ballin M, Griep W, Patel M, Karl M, Mentrup T, Rivera‐Monroy J, Foo B, Schwappach B, Schröder B. The intramembrane proteases
SPPL2a
and
SPPL2b
regulate the homeostasis of selected
SNARE
proteins. FEBS J 2022; 290:2320-2337. [PMID: 36047592 DOI: 10.1111/febs.16610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/28/2022] [Accepted: 08/30/2022] [Indexed: 01/15/2023]
Abstract
Signal peptide peptidase (SPP) and SPP-like (SPPL) aspartyl intramembrane proteases are known to contribute to sequential processing of type II-oriented membrane proteins referred to as regulated intramembrane proteolysis. The ER-resident family members SPP and SPPL2c were shown to also cleave tail-anchored proteins, including selected SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins facilitating membrane fusion events. Here, we analysed whether the related SPPL2a and SPPL2b proteases, which localise to the endocytic or late secretory pathway, are also able to process SNARE proteins. Therefore, we screened 18 SNARE proteins for cleavage by SPPL2a and SPPL2b based on cellular co-expression assays, of which the proteins VAMP1, VAMP2, VAMP3 and VAMP4 were processed by SPPL2a/b demonstrating the capability of these two proteases to proteolyse tail-anchored proteins. Cleavage of the four SNARE proteins was scrutinised at the endogenous level upon SPPL2a/b inhibition in different cell lines as well as by analysing VAMP1-4 levels in tissues and primary cells of SPPL2a/b double-deficient (dKO) mice. Loss of SPPL2a/b activity resulted in an accumulation of VAMP1-4 in a cell type- and tissue-dependent manner, identifying these proteins as SPPL2a/b substrates validated in vivo. Therefore, we propose that SPPL2a/b control cellular levels of VAMP1-4 by initiating the degradation of these proteins, which might impact cellular trafficking.
Collapse
Affiliation(s)
- Moritz Ballin
- Biochemical Institute Christian Albrechts University Kiel Kiel Germany
- Institute of Physiological Chemistry Technische Universität Dresden Dresden Germany
| | - Wolfram Griep
- Biochemical Institute Christian Albrechts University Kiel Kiel Germany
- Institute of Physiological Chemistry Technische Universität Dresden Dresden Germany
| | - Mehul Patel
- Institute of Physiological Chemistry Technische Universität Dresden Dresden Germany
| | - Martin Karl
- Institute of Physiological Chemistry Technische Universität Dresden Dresden Germany
| | - Torben Mentrup
- Institute of Physiological Chemistry Technische Universität Dresden Dresden Germany
| | - Jhon Rivera‐Monroy
- Department of Molecular Biology University Medical Center Göttingen Göttingen Germany
| | - Brian Foo
- Department of Molecular Biology University Medical Center Göttingen Göttingen Germany
| | - Blanche Schwappach
- Department of Molecular Biology University Medical Center Göttingen Göttingen Germany
| | - Bernd Schröder
- Institute of Physiological Chemistry Technische Universität Dresden Dresden Germany
| |
Collapse
|
122
|
Castañeyra-Ruiz L, González-Marrero I, Hernández-Abad LG, Lee S, Castañeyra-Perdomo A, Muhonen M. AQP4, Astrogenesis, and Hydrocephalus: A New Neurological Perspective. Int J Mol Sci 2022; 23:10438. [PMID: 36142348 PMCID: PMC9498986 DOI: 10.3390/ijms231810438] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Aquaporin 4 (AQP4) is a cerebral glial marker that labels ependymal cells and astrocytes' endfeet and is the main water channel responsible for the parenchymal fluid balance. However, in brain development, AQP4 is a marker of glial stem cells and plays a crucial role in the pathophysiology of pediatric hydrocephalus. Gliogenesis characterization has been hampered by a lack of biomarkers for precursor and intermediate stages and a deeper understanding of hydrocephalus etiology is needed. This manuscript is a focused review of the current research landscape on AQP4 as a possible biomarker for gliogenesis and its influence in pediatric hydrocephalus, emphasizing reactive astrogliosis. The goal is to understand brain development under hydrocephalic and normal physiologic conditions.
Collapse
Affiliation(s)
| | - Ibrahim González-Marrero
- Departamento de Ciencias Médicas Basicas, Anatomía, Facultad de Medicina, Universidad de La Laguna, Ofra s/n, 38071 La Laguna, Spain
| | - Luis G. Hernández-Abad
- Departamento de Ciencias Médicas Basicas, Anatomía, Facultad de Medicina, Universidad de La Laguna, Ofra s/n, 38071 La Laguna, Spain
| | - Seunghyun Lee
- CHOC Children’s Research Institute, 1201 W, La Veta Avenue, Orange, CA 92868, USA
| | - Agustín Castañeyra-Perdomo
- Departamento de Ciencias Médicas Basicas, Anatomía, Facultad de Medicina, Universidad de La Laguna, Ofra s/n, 38071 La Laguna, Spain
- Instituto de Investigación y Ciencias de Puerto del Rosario, 35600 Puerto del Rosario, Spain
| | - Michael Muhonen
- Neurosurgery Department at CHOC Children’s Hospital, 505 S Main St., Orange, CA 92868, USA
| |
Collapse
|
123
|
Castañeda-Sampedro A, Calvin-Cejudo L, Martin F, Gomez-Diaz C, Alcorta E. The Ntan1 gene is expressed in perineural glia and neurons of adult Drosophila. Sci Rep 2022; 12:14749. [PMID: 36042338 PMCID: PMC9427837 DOI: 10.1038/s41598-022-18999-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
The Drosophila Ntan1 gene encodes an N-terminal asparagine amidohydrolase that we show is highly conserved throughout evolution. Protein isoforms share more than 72% of similarity with their human counterparts. At the cellular level, this gene regulates the type of glial cell growth in Drosophila larvae by its different expression levels. The Drosophila Ntan1 gene has 4 transcripts that encode 2 protein isoforms. Here we describe that although this gene is expressed at all developmental stages and adult organs tested (eye, antennae and brain) there are some transcript-dependent specificities. Therefore, both quantitative and qualitative cues could account for gene function. However, widespread developmental stage and organ-dependent expression could be masking cell-specific constraints that can be explored in Drosophila by using Gal4 drivers. We report a new genetic driver within this gene, Mz317-Gal4, that recapitulates the Ntan1 gene expression pattern in adults. It shows specific expression for perineural glia in the olfactory organs but mixed expression with some neurons in the adult brain. Memory and social behavior disturbances in mice and cancer and schizophrenia in humans have been linked to the Ntan1 gene. Therefore, these new tools in Drosophila may contribute to our understanding of the cellular basis of these alterations.
Collapse
Affiliation(s)
- Ana Castañeda-Sampedro
- Facultad de Medicina y Ciencias de la Salud, Departamento de Biología Funcional (Área de Genética), Universidad de Oviedo, c/Julián Clavería S/N, 33006, Oviedo, Asturias, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Oviedo, Asturias, Spain
| | - Laura Calvin-Cejudo
- Facultad de Medicina y Ciencias de la Salud, Departamento de Biología Funcional (Área de Genética), Universidad de Oviedo, c/Julián Clavería S/N, 33006, Oviedo, Asturias, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Oviedo, Asturias, Spain
| | - Fernando Martin
- Facultad de Medicina y Ciencias de la Salud, Departamento de Biología Funcional (Área de Genética), Universidad de Oviedo, c/Julián Clavería S/N, 33006, Oviedo, Asturias, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Oviedo, Asturias, Spain
| | - Carolina Gomez-Diaz
- Facultad de Medicina y Ciencias de la Salud, Departamento de Biología Funcional (Área de Genética), Universidad de Oviedo, c/Julián Clavería S/N, 33006, Oviedo, Asturias, Spain. .,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Oviedo, Asturias, Spain.
| | - Esther Alcorta
- Facultad de Medicina y Ciencias de la Salud, Departamento de Biología Funcional (Área de Genética), Universidad de Oviedo, c/Julián Clavería S/N, 33006, Oviedo, Asturias, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Oviedo, Asturias, Spain
| |
Collapse
|
124
|
Huang H, He W, Tang T, Qiu M. Immunological Markers for Central Nervous System Glia. Neurosci Bull 2022; 39:379-392. [PMID: 36028641 PMCID: PMC10043115 DOI: 10.1007/s12264-022-00938-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/09/2022] [Indexed: 10/15/2022] Open
Abstract
Glial cells in the central nervous system (CNS) are composed of oligodendrocytes, astrocytes and microglia. They contribute more than half of the total cells of the CNS, and are essential for neural development and functioning. Studies on the fate specification, differentiation, and functional diversification of glial cells mainly rely on the proper use of cell- or stage-specific molecular markers. However, as cellular markers often exhibit different specificity and sensitivity, careful consideration must be given prior to their application to avoid possible confusion. Here, we provide an updated overview of a list of well-established immunological markers for the labeling of central glia, and discuss the cell-type specificity and stage dependency of their expression.
Collapse
Affiliation(s)
- Hao Huang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Wanjun He
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Tao Tang
- Department of Anatomy, Cell Biology and Physiology Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
125
|
Zhang J, Yang H, Wu J, Zhang D, Wang Y, Zhai J. Recent progresses in novel in vitro models of primary neurons: A biomaterial perspective. Front Bioeng Biotechnol 2022; 10:953031. [PMID: 36061442 PMCID: PMC9428288 DOI: 10.3389/fbioe.2022.953031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Central nervous system (CNS) diseases have been a growing threat to the health of humanity, emphasizing the urgent need of exploring the pathogenesis and therapeutic approaches of various CNS diseases. Primary neurons are directly obtained from animals or humans, which have wide applications including disease modeling, mechanism exploration and drug development. However, traditional two-dimensional (2D) monoculture cannot resemble the native microenvironment of CNS. With the increasing understanding of the complexity of the CNS and the remarkable development of novel biomaterials, in vitro models have experienced great innovation from 2D monoculture toward three-dimensional (3D) multicellular culture. The scope of this review includes the progress of various in vitro models of primary neurons in recent years to provide a holistic view of the modalities and applications of primary neuron models and how they have been connected with the revolution of biofabrication techniques. Special attention has been paid to the interaction between primary neurons and biomaterials. First, a brief introduction on the history of CNS modeling and primary neuron culture was conducted. Next, detailed progress in novel in vitro models were discussed ranging from 2D culture, ex vivo model, spheroid, scaffold-based model, 3D bioprinting model, and microfluidic chip. Modalities, applications, advantages, and limitations of the aforementioned models were described separately. Finally, we explored future prospects, providing new insights into how basic science research methodologies have advanced our understanding of the CNS, and highlighted some future directions of primary neuron culture in the next few decades.
Collapse
Affiliation(s)
- Jiangang Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huiyu Yang
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaming Wu
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dingyue Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiliang Zhai
- Departments of Orthopedics Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Jiliang Zhai,
| |
Collapse
|
126
|
Lichterfeld Y, Kalinski L, Schunk S, Schmakeit T, Feles S, Frett T, Herrmann H, Hemmersbach R, Liemersdorf C. Hypergravity Attenuates Reactivity in Primary Murine Astrocytes. Biomedicines 2022; 10:biomedicines10081966. [PMID: 36009513 PMCID: PMC9405820 DOI: 10.3390/biomedicines10081966] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/31/2022] [Accepted: 08/11/2022] [Indexed: 12/11/2022] Open
Abstract
Neuronal activity is the key modulator of nearly every aspect of behavior, affecting cognition, learning, and memory as well as motion. Hence, disturbances of the transmission of synaptic signals are the main cause of many neurological disorders. Lesions to nervous tissues are associated with phenotypic changes mediated by astrocytes becoming reactive. Reactive astrocytes form the basis of astrogliosis and glial scar formation. Astrocyte reactivity is often targeted to inhibit axon dystrophy and thus promote neuronal regeneration. Here, we aim to understand the impact of gravitational loading induced by hypergravity to potentially modify key features of astrocyte reactivity. We exposed primary murine astrocytes as a model system closely resembling the in vivo reactivity phenotype on custom-built centrifuges for cultivation as well as for live-cell imaging under hypergravity conditions in a physiological range (2g and 10g). We revealed spreading rates, migration velocities, and stellation to be diminished under 2g hypergravity. In contrast, proliferation and apoptosis rates were not affected. In particular, hypergravity attenuated reactivity induction. We observed cytoskeletal remodeling of actin filaments and microtubules under hypergravity. Hence, the reorganization of these key elements of cell structure demonstrates that fundamental mechanisms on shape and mobility of astrocytes are affected due to altered gravity conditions. In future experiments, potential target molecules for pharmacological interventions that attenuate astrocytic reactivity will be investigated. The ultimate goal is to enhance neuronal regeneration for novel therapeutic approaches.
Collapse
Affiliation(s)
- Yannick Lichterfeld
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Laura Kalinski
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Sarah Schunk
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Theresa Schmakeit
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Sebastian Feles
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Timo Frett
- Department of Muscle and Bone Metabolism, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Harald Herrmann
- Institute of Neuropathology, University of Erlangen, 91054 Erlangen, Germany
| | - Ruth Hemmersbach
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Christian Liemersdorf
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
- Correspondence: ; Tel.: +49-176-811-09-333
| |
Collapse
|
127
|
Zhang YH, Peng F, Zhang L, Kang K, Yang M, Chen C, Yu H. LONG NONCODING RNA UPREGULATES ADAPTER SHCA PROTEIN EXPRESSION TO PROMOTE COGNITIVE IMPAIRMENT AFTER CARDIAC ARREST AND RESUSCITATION. Shock 2022; 58:169-178. [PMID: 35953462 DOI: 10.1097/shk.0000000000001964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Aim: More patients are resuscitated from cardiac arrest and cardiopulmonary resuscitation (CA/CPR) due to advances in medical care. However, the burden now lies with post-cardiac arrest cognitive impairment in CA/CPR survivors. Based on our previous study, we aimed to further confirm the correlation between the long noncoding RNA-promoting ShcA (lncRNA-PS)/Src homology and collagen A (ShcA) axis and CA/CPR-induced cognitive impairment in molecular, cellular, and tissue levels. Methods and Results: The in vivo experiments were based on a mouse model of CA/CPR, while oxygen-glucose deprivation and reoxygenation was used as a cell model in vitro. Conditional ShcA suppression in neurons of the hippocampal CA1 region was achieved by cyclization recombinase of bacteriophage P1 recognizing DNA fragment locus of x-over P1 site (Cre/LoxP recombination system). Genetic manipulation of HT22 was achieved by lentivirus targeting lncRNA-PS and ShcA. Neurological function score was remarkably decreased, and cognitive function was affected after restoration of spontaneous circulation. LncRNA-PS and ShcA overexpression after CA/CPR, mainly happened in neurons of hippocampal CA1 region, was observed by in situ hybridization and immunofluorescence. Neuronal ShcA knockdown in hippocampal CA1 region before CA/CPR attenuated cognitive impairment after CA/CPR. ShcA deficiency protected HT22 cell line against oxygen-glucose deprivation and reoxygenation by inhibiting inflammation and apoptosis. In vitro upregulation of lncRNA-PS elevated ShcA expression, which was reversed by knockdown of ShcA. Conclusions: This study revealed that lncRNA-PS/ShcA axis is critically involved in the pathogenesis of cognitive impairment after CA/CPR. By inhibiting ShcA expression in neurons of the hippocampal CA1 region could improve the survival outcomes in mice after CA/CPR.
Collapse
|
128
|
Gorter RP, Baron W. Recent insights into astrocytes as therapeutic targets for demyelinating diseases. Curr Opin Pharmacol 2022; 65:102261. [PMID: 35809402 DOI: 10.1016/j.coph.2022.102261] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/19/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023]
Abstract
Astrocytes are a group of glial cells that exhibit great morphological, transcriptional and functional diversity both in the resting brain and in response to injury. In recent years, astrocytes have attracted increasing interest as therapeutic targets for demyelinating diseases. Following a demyelinating insult, astrocytes can adopt a wide spectrum of reactive states, which can exacerbate damage, but may also facilitate oligodendrocyte progenitor cell differentiation and myelin regeneration. In this review, we provide an overview of recent literature on astrocyte-oligodendrocyte interactions in the context of demyelinating diseases. We highlight novel key roles for astrocytes both during demyelination and remyelination with a focus on potential therapeutic strategies to favor a pro-regenerative astrocyte response in (progressive) multiple sclerosis.
Collapse
Affiliation(s)
- Rianne Petra 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
|
129
|
Huang B, He Y, Rofaani E, Liang F, Huang X, Shi J, Wang L, Yamada A, Peng J, Chen Y. Automatic differentiation of human induced pluripotent stem cells toward synchronous neural networks on an arrayed monolayer of nanofiber membrane. Acta Biomater 2022; 150:168-180. [PMID: 35907558 DOI: 10.1016/j.actbio.2022.07.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/01/2022] [Accepted: 07/21/2022] [Indexed: 11/01/2022]
Abstract
Automatic differentiation of human-induced pluripotent stem cells (hiPSCs) facilitates the generation of cortical neural networks and studies of brain functions. Here, we present a method of directed differentiation of hiPSCs with a substrate made of a honeycomb microframe and a monolayer of crosslinked gelatin nanofibers in the form of an array of nanofiber membranes. Neural precursor cells (NPCs) were firstly derived from hiPSCs and then placed on the nanofiber membranes for automatically controlled neural differentiation over a long period. Due to the strong modulation of the substrate stiffness and permeability, most cells were found in the center area of the honeycomb compartments, giving rise to regular and inter-connected cortical neural clusters. More importantly, the neural activities of the clusters were synchronized proving the reliability of the method. Our results showed that the self-organization, as well as the neural activities of differentiating neural cells, were more efficient in the nanofiber membrane compared to the types of the substrate such as glass and nanofiber-covered glass. In addition to the inherent advantages such as manpower saving and fewer risks of contamination and human error, automatic differentiation avoided undesired shaking which might have critical effects on the formation of synchronous neural clusters. STATEMENT OF SIGNIFICANCE: : Synchronization of cortical neural activities is essential for information processing and human cognition. By automated differentiation of human induced pluripotent stem cells on arrayed monolayer of nanofiber membrane, synchronous neural clusters could be formed. Such an approach would allow creating a variety of neural networks with regular and interconnected clusters for systematic studies of human cortical functions.
Collapse
Affiliation(s)
- Boxin Huang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Yong He
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Elrade Rofaani
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Feng Liang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Xiaochen Huang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Jian Shi
- MesoBioTech, 231 Rue Saint-Honoré, 75001, Paris, France
| | - Li Wang
- MesoBioTech, 231 Rue Saint-Honoré, 75001, Paris, France
| | - Ayako Yamada
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Juan Peng
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| | - Yong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| |
Collapse
|
130
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
131
|
Paton KM, Selfridge J, Guy J, Bird A. Comparative analysis of potential broad-spectrum neuronal Cre drivers. Wellcome Open Res 2022; 7:185. [PMID: 35966957 PMCID: PMC9353198 DOI: 10.12688/wellcomeopenres.17965.1] [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] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
Cre/Lox technology is a powerful tool in the mouse genetics tool-box as it enables tissue-specific and inducible mutagenesis of specific gene loci. Correct interpretation of phenotypes depends upon knowledge of the Cre expression pattern in the chosen mouse driver line to ensure that appropriate cell types are targeted. For studies of the brain and neurological disease a pan-neuronal promoter that reliably drives efficient neuron-specific transgene expression would be valuable. Here we compare a widely used "pan-neuronal" mouse Cre driver line, Syn1-cre, with a little-known alternative, Snap25-IRES2-cre. Our results show that the Syn1-cre line broadly expresses in the brain but is indetectable in more than half of all neurons and weakly active in testes. In contrast the Snap25-IRES2-cre line expressed Cre in a high proportion of neurons (~85%) and was indetectable in all non-brain tissues that were analysed, including testes. Our findings suggest that for many purposes Snap25-IRES2-cre is superior to Syn1-cre as a potential pan-neuronal cre driver.
Collapse
Affiliation(s)
- Katie M Paton
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Jim Selfridge
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Jacky Guy
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Adrian Bird
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| |
Collapse
|
132
|
Ventura-Antunes L, Dasgupta OM, Herculano-Houzel S. Resting Rates of Blood Flow and Glucose Use per Neuron Are Proportional to Number of Endothelial Cells Available per Neuron Across Sites in the Rat Brain. Front Integr Neurosci 2022; 16:821850. [PMID: 35757100 PMCID: PMC9226568 DOI: 10.3389/fnint.2022.821850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/07/2022] [Indexed: 11/24/2022] Open
Abstract
We report in a companion paper that in the mouse brain, in contrast to the 1,000-fold variation in local neuronal densities across sites, capillary density (measured both as capillary volume fraction and as density of endothelial cells) show very little variation, of the order of only fourfold. Here we confirm that finding in the rat brain and, using published rates of local blood flow and glucose use at rest, proceed to show that what small variation exists in capillary density across sites in the rat brain is strongly and linearly correlated to variations in local rates of brain metabolism at rest. Crucially, we show that such variations in local capillary density and brain metabolism are not correlated with local variations in neuronal density, which contradicts expectations that use-dependent self-organization would cause brain sites with more neurons to have higher capillary densities due to higher energetic demands. In fact, we show that the ratio of endothelial cells per neuron serves as a linear indicator of average blood flow and glucose use per neuron at rest, and both increase as neuronal density decreases across sites. In other words, because of the relatively tiny variation in capillary densities compared to the large variation in neuronal densities, the anatomical infrastructure of the brain is such that those sites with fewer neurons have more energy supplied per neuron, which matches a higher average rate of energy use per neuron, compared to sites with more neurons. Taken together, our data support the interpretation that resting brain metabolism is not demand-based, but rather limited by its capillary supply, and raise multiple implications for the differential vulnerability of diverse brain areas to disease and aging.
Collapse
Affiliation(s)
- Lissa Ventura-Antunes
- Department of Psychology, Vanderbilt University, Nashville, TN, United States.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | | | - Suzana Herculano-Houzel
- Department of Psychology, Vanderbilt University, Nashville, TN, United States.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
133
|
Ortinski PI, Reissner KJ, Turner J, Anderson TA, Scimemi A. Control of complex behavior by astrocytes and microglia. Neurosci Biobehav Rev 2022; 137:104651. [PMID: 35367512 PMCID: PMC9119927 DOI: 10.1016/j.neubiorev.2022.104651] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/28/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Evidence that glial cells influence behavior has been gaining a steady foothold in scientific literature. Out of the five main subtypes of glial cells in the brain, astrocytes and microglia have received an outsized share of attention with regard to shaping a wide spectrum of behavioral phenomena and there is growing appreciation that the signals intrinsic to these cells as well as their interactions with surrounding neurons reflect behavioral history in a brain region-specific manner. Considerable regional diversity of glial cell phenotypes is beginning to be recognized and may contribute to behavioral outcomes arising from circuit-specific computations within and across discrete brain nuclei. Here, we summarize current knowledge on the impact of astrocyte and microglia activity on behavioral outcomes, with a specific focus on brain areas relevant to higher cognitive control, reward-seeking, and circadian regulation.
Collapse
Affiliation(s)
- P I Ortinski
- Department of Neuroscience, University of Kentucky, USA
| | - K J Reissner
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, USA
| | - J Turner
- Department of Pharmaceutical Sciences, University of Kentucky, USA
| | - T A Anderson
- Department of Neuroscience, University of Kentucky, USA
| | - A Scimemi
- Department of Biology, State University of New York at Albany, USA
| |
Collapse
|
134
|
Han G, Bai K, Yang X, Sun C, Ji Y, Zhou J, Zhang H, Ding Y. "Drug-Carrier" Synergy Therapy for Amyloid-β Clearance and Inhibition of Tau Phosphorylation via Biomimetic Lipid Nanocomposite Assembly. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2106072. [PMID: 35307993 PMCID: PMC9108666 DOI: 10.1002/advs.202106072] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/19/2022] [Indexed: 05/13/2023]
Abstract
Amyloid-β (Aβ) toxicity is considered to be companioned by Tau phosphorylation in Alzheimer's disease (AD). The clinical AD therapy is usually subjected to low blood-brain barrier (BBB) penetration and complex interaction mechanisms between Aβ and phosphorylated Tau. A "Drug-Carrier" synergy therapy is herein designed to simultaneously target Aβ and Tau-associated pathways for AD treatment. To imitate natural nanoparticle configuration, the endogenous apolipoprotein A-I and its mimicking peptide 4F fused angiopep-2 (Ang) are sequentially grafted onto lipid nanocomposite (APLN), providing liberty of BBB crossing and microglia targeted Aβ clearance. For synergy treatment, methylene blue (MB) is further assembled into APLN (APLN/MB) for Tau aggregation inhibition. After intravenous administration, the optimized density (5 wt%) of Ang ligands dramatically enhances APLN/MB intracerebral shuttling and accumulation, which is 2.15-fold higher than that Ang absent-modification. The site-specific release of MB collaborates APLN to promote Aβ capture for microglia endocytosis clearance and reduce p-Tau level by 25.31% in AD pathogenesis. In AD-Aβ-Tau bearing mouse models, APLN/MB can relieve AD symptoms, rescue neuron viability and cognitive functions. Collectively, it is confirmed that "Drug-Carrier" synergy therapy of APLN/MB is a promising approach in the development of AD treatments.
Collapse
Affiliation(s)
- Guochen Han
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Kaiwen Bai
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Xiaoyu Yang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Chenhua Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Yi Ji
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Jianping Zhou
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Yang Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)State Key Laboratory of Natural MedicinesDepartment of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| |
Collapse
|
135
|
Schneider J, Weigel J, Wittmann MT, Svehla P, Ehrt S, Zheng F, Elmzzahi T, Karpf J, Paniagua-Herranz L, Basak O, Ekici A, Reis A, Alzheimer C, Ortega de la O F, Liebscher S, Beckervordersandforth R. Astrogenesis in the murine dentate gyrus is a life-long and dynamic process. EMBO J 2022; 41:e110409. [PMID: 35451150 PMCID: PMC9156974 DOI: 10.15252/embj.2021110409] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are highly abundant in the mammalian brain, and their functions are of vital importance for all aspects of development, adaption, and aging of the central nervous system (CNS). Mounting evidence indicates the important contributions of astrocytes to a wide range of neuropathies. Still, our understanding of astrocyte development significantly lags behind that of other CNS cells. We here combine immunohistochemical approaches with genetic fate-mapping, behavioral paradigms, single-cell transcriptomics, and in vivo two-photon imaging, to comprehensively assess the generation and the proliferation of astrocytes in the dentate gyrus (DG) across the life span of a mouse. Astrogenesis in the DG is initiated by radial glia-like neural stem cells giving rise to locally dividing astrocytes that enlarge the astrocyte compartment in an outside-in-pattern. Also in the adult DG, the vast majority of astrogenesis is mediated through the proliferation of local astrocytes. Interestingly, locally dividing astrocytes were able to adapt their proliferation to environmental and behavioral stimuli revealing an unexpected plasticity. Our study establishes astrocytes as enduring plastic elements in DG circuits, implicating a vital contribution of astrocyte dynamics to hippocampal plasticity.
Collapse
Affiliation(s)
- Julia Schneider
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Weigel
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marie-Theres Wittmann
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Pavel Svehla
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians University Munich, Munich, Germany.,Medical Faculty, BioMedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Sebastian Ehrt
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany.,Medical Faculty, BioMedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tarek Elmzzahi
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Molecular Immunology in Neurodegeneration, German Centre for Neurodegenerative Diseases Bonn, Bonn, Germany
| | - Julian Karpf
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lucía Paniagua-Herranz
- Department of Molecular Biology, Universidad Complutense de Madrid, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Sanitaria San Carlos (IdISSC), Spain
| | - Onur Basak
- Department of Translational Neuroscience, University Medical Centre Utrecht (UMCU), Utrecht, Netherlands
| | - Arif Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andre Reis
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Felipe Ortega de la O
- Department of Molecular Biology, Universidad Complutense de Madrid, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Sanitaria San Carlos (IdISSC), Spain
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians University Munich, Munich, Germany.,Medical Faculty, BioMedical Center, Ludwig-Maximilians University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | |
Collapse
|
136
|
Lukow PB, Martins D, Veronese M, Vernon AC, McGuire P, Turkheimer FE, Modinos G. Cellular and molecular signatures of in vivo imaging measures of GABAergic neurotransmission in the human brain. Commun Biol 2022; 5:372. [PMID: 35440709 PMCID: PMC9018713 DOI: 10.1038/s42003-022-03268-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
Diverse GABAergic interneuron networks orchestrate information processing in the brain. Understanding the principles underlying the organisation of this system in the human brain, and whether these principles are reflected by available non-invasive in vivo neuroimaging methods, is crucial for the study of GABAergic neurotransmission. Here, we use human gene expression data and state-of-the-art imaging transcriptomics to uncover co-expression patterns between genes encoding GABAA receptor subunits and inhibitory interneuron subtype-specific markers, and their association with binding patterns of the gold-standard GABA PET radiotracers [11C]Ro15-4513 and [11C]flumazenil. We found that the inhibitory interneuron marker somatostatin covaries with GABAA receptor-subunit genes GABRA5 and GABRA2, and that their distribution followed [11C]Ro15-4513 binding. In contrast, the inhibitory interneuron marker parvalbumin covaried with GABAA receptor-subunit genes GABRA1, GABRB2 and GABRG2, and their distribution tracked [11C]flumazenil binding. Our findings indicate that existing PET radiotracers may provide complementary information about key components of the GABAergic system.
Collapse
Affiliation(s)
- Paulina Barbara Lukow
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK.
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- NIHR Maudsley Biomedical Research Centre, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- NIHR Maudsley Biomedical Research Centre, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
- Department of Information Engineering, University of Padua, Via Giovanni Gradenigo, 6, 35131, Padova, PD, Italy
| | - Anthony Christopher Vernon
- Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, Brixton, London, SE5 9RT, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- NIHR Maudsley Biomedical Research Centre, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - Federico Edoardo Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
| | - Gemma Modinos
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, New Hunt's House, Guy's Campus, London, UK
| |
Collapse
|
137
|
Cathomas F, Holt LM, Parise EM, Liu J, Murrough JW, Casaccia P, Nestler EJ, Russo SJ. Beyond the neuron: Role of non-neuronal cells in stress disorders. Neuron 2022; 110:1116-1138. [PMID: 35182484 PMCID: PMC8989648 DOI: 10.1016/j.neuron.2022.01.033] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Stress disorders are leading causes of disease burden in the U.S. and worldwide, yet available therapies are fully effective in less than half of all individuals with these disorders. Although to date, much of the focus has been on neuron-intrinsic mechanisms, emerging evidence suggests that chronic stress can affect a wide range of cell types in the brain and periphery, which are linked to maladaptive behavioral outcomes. Here, we synthesize emerging literature and discuss mechanisms of how non-neuronal cells in limbic regions of brain interface at synapses, the neurovascular unit, and other sites of intercellular communication to mediate the deleterious, or adaptive (i.e., pro-resilient), effects of chronic stress in rodent models and in human stress-related disorders. We believe that such an approach may one day allow us to adopt a holistic "whole body" approach to stress disorder research, which could lead to more precise diagnostic tests and personalized treatment strategies. Stress is a major risk factor for many psychiatric disorders. Cathomas et al. review new insight into how non-neuronal cells mediate the deleterious effects, as well as the adaptive, protective effects, of stress in rodent models and human stress-related disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanne M Holt
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
138
|
Mou W, Ma L, Zhu A, Cui H, Huang Y. Astrocyte-microglia interaction through C3/C3aR pathway modulates neuropathic pain in rats model of chronic constriction injury. Mol Pain 2022; 18:17448069221140532. [PMID: 36341694 PMCID: PMC9669679 DOI: 10.1177/17448069221140532] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 09/24/2022] [Accepted: 11/03/2022] [Indexed: 12/30/2023] Open
Abstract
Neuropathic pain (NP) is the cardinal symptom of neural injury, and its underlying molecular mechanism needs further investigation. Complements, especially complement 3 (C3), are involved in the pathophysiology of many neurological disorders, while the specific role of C3 in NP is still obscure. In this study, we found that both C3 and its receptor C3aR were upregulated in the spinal dorsal horn in a rat chronic constriction injury (CCI) model. In addition, C3 was mainly detected in astrocytes, while C3aR was expressed in microglia and neuron. Intrathecal injection of C3 antibody and C3aR antagonist alleviated NP in CCI model together with reduced M1 polarization of microglia. Our finding suggested that blockade of the C3/C3aR pathway might be a novel strategy for NP.
Collapse
Affiliation(s)
- Wanying Mou
- Department of Anesthesiology,
Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College
Hospital, Beijing, China
| | - Lulu Ma
- Department of Anesthesiology,
Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College
Hospital, Beijing, China
| | - Afang Zhu
- Department of Anesthesiology,
Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College
Hospital, Beijing, China
| | - Huan Cui
- Department of Human Anatomy,
Histology and Embryology, Neuroscience Center, Institute of Basic Medical
Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical
College, Beijing, China
| | - Yuguang Huang
- Department of Anesthesiology,
Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College
Hospital, Beijing, China
| |
Collapse
|
139
|
Irie K, Doi M, Usui N, Shimada S. Evolution of the Human Brain Can Help Determine Pathophysiology of Neurodevelopmental Disorders. Front Neurosci 2022; 16:871979. [PMID: 35431788 PMCID: PMC9010664 DOI: 10.3389/fnins.2022.871979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/28/2022] [Indexed: 02/03/2023] Open
Abstract
The evolution of humans brought about a co-occurring evolution of the human brain, which is far larger and more complex than that of many other organisms. The brain has evolved characteristically in humans in many respects, including macro-and micro-anatomical changes in the brain structure, changes in gene expression, and cell populations and ratios. These characteristics are essential for the execution of higher functions, such as sociality, language, and cognition, which express humanity, and are thought to have been acquired over evolutionary time. However, with the acquisition of higher functions also comes the risk of the disease in which they fail. This review focuses on human brain evolution and neurodevelopmental disorders (NDDs) and discusses brain development, molecular evolution, and human brain evolution. Discussing the potential for the development and pathophysiology of NDDs acquired by human brain evolution will provide insights into the acquisition and breakdown of higher functions from a new perspective.
Collapse
Affiliation(s)
- Koichiro Irie
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- Center for Medical Research and Education, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Miyuki Doi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- United Graduate School of Child Development, Osaka University, Suita, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
- *Correspondence: Noriyoshi Usui,
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- United Graduate School of Child Development, Osaka University, Suita, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| |
Collapse
|
140
|
Beeraka NM, Vikram PRH, Greeshma MV, Uthaiah CA, Huria T, Liu J, Kumar P, Nikolenko VN, Bulygin KV, Sinelnikov MY, Sukocheva O, Fan R. Recent Investigations on Neurotransmitters' Role in Acute White Matter Injury of Perinatal Glia and Pharmacotherapies-Glia Dynamics in Stem Cell Therapy. Mol Neurobiol 2022; 59:2009-2026. [PMID: 35041139 DOI: 10.1007/s12035-021-02700-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023]
Abstract
Periventricular leukomalacia (PVL) and cerebral palsy are two neurological disease conditions developed from the premyelinated white matter ischemic injury (WMI). The significant pathophysiology of these diseases is accompanied by the cognitive deficits due to the loss of function of glial cells and axons. White matter makes up 50% of the brain volume consisting of myelinated and non-myelinated axons, glia, blood vessels, optic nerves, and corpus callosum. Studies over the years have delineated the susceptibility of white matter towards ischemic injury especially during pregnancy (prenatal, perinatal) or immediately after child birth (postnatal). Impairment in membrane depolarization of neurons and glial cells by ischemia-invoked excitotoxicity is mediated through the overactivation of NMDA receptors or non-NMDA receptors by excessive glutamate influx, calcium, or ROS overload and has been some of the well-studied molecular mechanisms conducive to the injury of white matter. Expression of glutamate receptors (GluR) and transporters (GLT1, EACC1, and GST) has significant influence in glial and axonal-mediated injury of premyelinated white matter during PVL and cerebral palsy. Predominantly, the central premyelinated axons express extensive levels of functional NMDA GluR receptors to confer ischemic injury to premyelinated white matter which in turn invoke defects in neural plasticity. Several underlying molecular mechanisms are yet to be unraveled to delineate the complete pathophysiology of these prenatal neurological diseases for developing the novel therapeutic modalities to mitigate pathophysiology and premature mortality of newborn babies. In this review, we have substantially discussed the above multiple pathophysiological aspects of white matter injury along with glial dynamics, and the pharmacotherapies including recent insights into the application of MSCs as therapeutic modality in treating white matter injury.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - P R Hemanth Vikram
- Department of Pharmaceutical Chemistry, JSS Pharmacy College, Mysuru, Karnataka, India
| | - M V Greeshma
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Chinnappa A Uthaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Tahani Huria
- Faculty of Medicine, Benghazi University, Benghazi, Libya
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, LE1 7RH, UK
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), SilaKatamur (Halugurisuk), Changsari, Kamrup, 781101, Assam, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Department of Normal and Topographic Anatomy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russian Federation
| | - Olga Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China.
| |
Collapse
|
141
|
Singh R, Bartok A, Paillard M, Tyburski A, Elliott M, Hajnóczky G. Uncontrolled mitochondrial calcium uptake underlies the pathogenesis of neurodegeneration in MICU1-deficient mice and patients. SCIENCE ADVANCES 2022; 8:eabj4716. [PMID: 35302860 PMCID: PMC8932652 DOI: 10.1126/sciadv.abj4716] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/26/2022] [Indexed: 06/01/2023]
Abstract
Dysregulation of mitochondrial Ca2+ homeostasis has been linked to neurodegenerative diseases. Mitochondrial Ca2+ uptake is mediated via the calcium uniporter complex that is primarily regulated by MICU1, a Ca2+-sensing gatekeeper. Recently, human patients with MICU1 loss-of-function mutations were diagnosed with neuromuscular and cognitive impairments. While studies in patient-derived cells revealed altered mitochondrial calcium signaling, the neuronal pathogenesis was difficult to study. To fill this void, we created a neuron-specific MICU1-KO mouse model. These animals show progressive, abnormal motor and cognitive phenotypes likely caused by the degeneration of motor neurons in the spinal cord and the cortex. We found increased susceptibility to mitochondrial Ca2+ overload-induced excitotoxic insults and cell death in MICU1-KO neurons and MICU1-deficient patient-derived cells, which can be blunted by inhibiting the mitochondrial permeability transition pore. Thus, our study identifies altered neuronal mitochondrial Ca2+ homeostasis as causative in the clinical symptoms of MICU1-deficient patients and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Raghavendra Singh
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Bartok
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Departent of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Melanie Paillard
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ashley Tyburski
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Melanie Elliott
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
142
|
Shinmyo Y, Saito K, Hamabe-Horiike T, Kameya N, Ando A, Kawasaki K, Duong TAD, Sakashita M, Roboon J, Hattori T, Kannon T, Hosomichi K, Slezak M, Holt MG, Tajima A, Hori O, Kawasaki H. Localized astrogenesis regulates gyrification of the cerebral cortex. SCIENCE ADVANCES 2022; 8:eabi5209. [PMID: 35275722 PMCID: PMC8916738 DOI: 10.1126/sciadv.abi5209] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
The development and evolution of mammalian higher cognition are represented by gyrification of the laminar cerebral cortex and astrocyte development, but their mechanisms and interrelationships remain unknown. Here, we show that localized astrogenesis plays an important role in gyri formation in the gyrencephalic cerebral cortex. In functional genetic experiments, we show that reducing astrocyte number prevents gyri formation in the ferret cortex, while increasing astrocyte number in mice, which do not have cortical folds, can induce gyrus-like protrusions. Morphometric analyses demonstrate that the vertical expansion of deep pallial regions achieved by localized astrogenesis is crucial for gyri formation. Furthermore, our findings suggest that localized astrogenesis by a positive feedback loop of FGF signaling is an important mechanism underlying cortical folding in gyrencephalic mammalian brains. Our findings reveal both the cellular mechanisms and the mechanical principle of gyrification in the mammalian brain.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Narufumi Kameya
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Akitaka Ando
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kanji Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Tung Anh Dinh Duong
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Masataka Sakashita
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Takayuki Kannon
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Michal Slezak
- VIB Center for Brain and Disease Research, Herestraat 49, Leuven 3000, Belgium
- Łukasiewicz Research Network-PORT Polish Institute for Technology Development, 54-066 Wroclaw, Poland
| | - Matthew G. Holt
- VIB Center for Brain and Disease Research, Herestraat 49, Leuven 3000, Belgium
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| |
Collapse
|
143
|
El-Nabulsi RA. Emergence of lump-like solitonic waves in Heimburg-Jackson biomembranes and nerves fractal model. J R Soc Interface 2022; 19:20220079. [PMID: 35317648 PMCID: PMC8941413 DOI: 10.1098/rsif.2022.0079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
The aim of this study is to extend the soliton propagation model in biomembranes and nerves constructed by Heimburg and Jackson for the case of fractal dimensions. Our analyses are based on the product-like fractal measure concept introduced by Li and Ostoja-Starzewski in their attempt to explore anisotropic fractal elastic media and electromagnetic fields. The mathematical model presented in the paper is formulated for only a part of a single nerve cell (an axon). The analytical and numerical envelop soliton of this equation are reported. The results obtained prove the emergence of lump-type solitonic waves in nerves and biomembranes. In particular, these waves decay algebraically to the background wave in space direction. This scenario is viewed as a particular class of rational localized waves which are solutions of the integrable Ishimori I equation and the (2 + 1) Kadomtsev-Petviashvili I equation. The effects of fractal dimensions are discussed and were found to be significant to some extents.
Collapse
Affiliation(s)
- Rami Ahmad El-Nabulsi
- Research Center for Quantum Technology, Faculty of Science, Chiang Mai University, 50200, Thailand
- Department of Physics and Materials Science, Faculty of Science, Chiang Mai University, 50200, Thailand
- Athens Institute for Education and Research, Mathematics and Physics Divisions, 8 Valaoritou Street, Kolonaki, 10671 Athens, Greece
| |
Collapse
|
144
|
Baudon A, Charlet A. [Reaching for the stars: How astrocytes regulate positive emotions via oxytocin]. Med Sci (Paris) 2022; 38:138-140. [PMID: 35179466 DOI: 10.1051/medsci/2021255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Angel Baudon
- CNRS et Université de Strasbourg, Institut des neurosciences cellulaires et intégratives (INCI, UPR3212), 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Alexandre Charlet
- CNRS et Université de Strasbourg, Institut des neurosciences cellulaires et intégratives (INCI, UPR3212), 8 allée du Général Rouvillois, 67000 Strasbourg, France
| |
Collapse
|
145
|
Reis PA, Castro-Faria-Neto HC. Systemic Response to Infection Induces Long-Term Cognitive Decline: Neuroinflammation and Oxidative Stress as Therapeutical Targets. Front Neurosci 2022; 15:742158. [PMID: 35250433 PMCID: PMC8895724 DOI: 10.3389/fnins.2021.742158] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/31/2021] [Indexed: 12/29/2022] Open
Abstract
In response to pathogens or damage signs, the immune system is activated in order to eliminate the noxious stimuli. The inflammatory response to infectious diseases induces systemic events, including cytokine storm phenomenon, vascular dysfunction, and coagulopathy, that can lead to multiple-organ dysfunction. The central nervous system (CNS) is one of the major organs affected, and symptoms such as sickness behavior (depression and fever, among others), or even delirium, can be observed due to activation of endothelial and glial cells, leading to neuroinflammation. Several reports have been shown that, due to CNS alterations caused by neuroinflammation, some sequels can be developed in special cognitive decline. There is still no any treatment to avoid cognitive impairment, especially those developed due to systemic infectious diseases, but preclinical and clinical trials have pointed out controlling neuroinflammatory events to avoid the development of this sequel. In this minireview, we point to the possible mechanisms that triggers long-term cognitive decline, proposing the acute neuroinflammatory events as a potential therapeutical target to treat this sequel that has been associated to several infectious diseases, such as malaria, sepsis, and, more recently, the new SARS-Cov2 infection.
Collapse
Affiliation(s)
- Patricia Alves Reis
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Biochemistry Department, Roberto Alcântara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Alves Reis,
| | | |
Collapse
|
146
|
Pawluski JL, Hoekzema E, Leuner B, Lonstein JS. Less can be more: Fine tuning the maternal brain. Neurosci Biobehav Rev 2022; 133:104475. [PMID: 34864004 PMCID: PMC8807930 DOI: 10.1016/j.neubiorev.2021.11.045] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 02/03/2023]
Abstract
PAWLUSKI, J.L., Hoekzema, E., Leuner, B., and Lonstein, J.S. Less can be more: Fine tuning the maternal brain. NEUROSCI BIOBEHAV REV (129) XXX-XXX, 2022. Plasticity in the female brain across the lifespan has recently become a growing field of scientific inquiry. This has led to the understanding that the transition to motherhood is marked by some of the most significant changes in brain plasticity in the adult female brain. Perhaps unexpectedly, plasticity occurring in the maternal brain often involves a decrease in brain volume, neurogenesis and glial cell density that presumably optimizes caregiving and other postpartum behaviors. This review summarizes what we know of the 'fine-tuning' of the female brain that accompanies motherhood and highlights the implications of these changes for maternal neurobehavioral health. The first part of the review summarizes structural and functional brain changes in humans during pregnancy and postpartum period with the remainder of the review focusing on neural and glial plasticity during the peripartum period in animal models. The aim of this review is to provide a clear understanding of when 'less is more' in maternal brain plasticity and where future research can focus to improve our understanding of the unique brain plasticity occurring during matrescence.
Collapse
Affiliation(s)
- Jodi L. Pawluski
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.,Corresponding author: Jodi L. Pawluski, University of Rennes 1, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| | - Elseline Hoekzema
- Brain and Development Laboratory, Leiden Institute for Brain and Cognition (LIBC), Leiden University, Leiden, The Netherlands.,Hoekzema Lab, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology & Department of Neuroscience Columbus, OH, USA
| | - Joseph S. Lonstein
- Neuroscience Program & Department of Psychology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
147
|
Nieder C, Rosene DL, Mortazavi F, Oblak AL, Ketten DR. Morphology and unbiased stereology of the lateral superior olive in the short‐beaked common dolphin,
Delphinus delphis
(Cetacea, Delphinidae). J Morphol 2022; 283:446-461. [DOI: 10.1002/jmor.21453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/10/2022] [Accepted: 01/16/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Carolin Nieder
- Institute of Marine Science, University of Auckland, Leigh Marine Laboratory, 160 Goat Island Road, Leigh New Zealand
| | - Douglas L. Rosene
- Department of Anatomy and Neurobiology Boston University School of Medicine 72 East, Concord St (L 1004), Boston Massachusetts
| | - Farzad Mortazavi
- Department of Anatomy and Neurobiology Boston University School of Medicine 72 East, Concord St (L 1004), Boston Massachusetts
| | - Adrian L. Oblak
- Indiana University School of Medicine, Stark Neurosciences Research Institute, Department of Radiology & Imaging Sciences, 320 W. 15th Street Indianapolis IN
| | - Darlene R. Ketten
- Woods Hole Oceanographic Institution, Biology Department, Marine Research Facility, MS #50 Woods Hole MA USA
| |
Collapse
|
148
|
Artificial Light at Night, Higher Brain Functions and Associated Neuronal Changes: An Avian Perspective. BIRDS 2022. [DOI: 10.3390/birds3010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent times, there has been an unprecedented increase in usage of electrical lightning. This has led to increase in artificial light at night (ALAN), and it has been suggested as a source of environmental pollution. ALAN exposure has been reported to be associated with disruption of daily rhythms and serious health consequences, such as immune, metabolic, and cognitive dysfunctions in both birds and mammals. Given the worldwide pervasiveness of ALAN, this research topic is also important from an ecological perspective. In birds, daily timings and appropriate temporal niches are important for fitness and survival. Daily rhythms in a wide array of functions are regulated by the circadian clock(s) and endogenous oscillators present in the body. There is accumulating evidence that exposure to ALAN disrupts clock-regulated daily rhythms and suppresses melatonin and sleep in birds. Circadian clock, melatonin, and sleep regulate avian cognitive performance. However, there is limited research on this topic, and most of the insights on the adverse effects of ALAN on cognitive functions are from behavioural studies. Nevertheless, these results raise an intriguing question about the molecular underpinning of the ALAN-induced negative consequences on brain functions. Further research should be focused on the molecular links between ALAN and cognitive performance, including the role of melatonin, which could shed light on the mechanism by which ALAN exposures lead to negative consequences.
Collapse
|
149
|
Unraveling unique and common cell type-specific mechanisms in glioblastoma multiforme. Comput Struct Biotechnol J 2022; 20:90-106. [PMID: 34976314 PMCID: PMC8688884 DOI: 10.1016/j.csbj.2021.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma multiforme persists to be an enigmatic distress in neuro-oncology. Its untethering capacity to thrive in a confined microenvironment, metastasize intracranially, and remain resistant to the systemic treatments, renders this tumour incurable. The glial cell type specificity in GBM remains exploratory. In our study, we aimed to address this problem by studying the GBM at the cell type level in the brain. The cellular makeup of this tumour is composed of genetically altered glial cells which include astrocyte, microglia, oligodendrocyte precursor cell, newly formed oligodendrocyte and myelinating oligodendrocyte. We extracted cell type-specific solid tumour as well as recurrent solid tumour glioma genes, and studied their functional networks and contribution towards gliomagenesis. We identified the principal transcription factors that are found to be regulating vital tumorigenic processes. We also assessed the protein-protein interaction networks at their domain level to get a more microscopic view of the structural and functional operations that transpire in these cells. This yielded the eminent protein regulators exhibiting their regulation in signaling pathways. Overall, our study unveiled regulatory mechanisms in glioma cell types that can be targeted for a more efficient glioma therapy.
Collapse
Key Words
- CAMs, Cell adhesion molecules
- CNS, Cental nervous system
- DEG, Differentially expressed genes
- EMT, Epithelial-mesenchymal transistion
- GBM, Glioblastoma multiforme
- GSC, Glioblastoma Stem Cell
- Glial cell types
- Glioblastoma multiforme
- INstruct, a database of structurally resolved protein interactome
- MO, Myelinating oligodendrocyte
- NCBI, National Centre for Biotechnology Information
- NFO, Newly formed oligodendrocyte
- NPC, Neural progenitor cell
- OPC, Oligodendrocyte precursor cell
- PDI, Protein domain interactions
- PDIN, Protein domain interaction network
- PPI, Protein-protein interactions
- Primary solid tumour
- Protein domains
- Protein interaction networks
- RSEM, RNA-seq by Expectation-Maximization
- Recurrent solid tumour transcription factors
- SIGNOR, Signaling Network Open Resource
- TCGA, The Cancer Genome Atlas
- TF, Transcription factor
- TP, Primary solid tumour
- TR, Recurrent solid tumour
- WHO, World health organization
- iDEP, Integrated Differential Expression and Pathway analysis
Collapse
|
150
|
Samanipour R, Tahmooressi H, Rezaei Nejad H, Hirano M, Shin SR, Hoorfar M. A review on 3D printing functional brain model. BIOMICROFLUIDICS 2022; 16:011501. [PMID: 35145569 PMCID: PMC8816519 DOI: 10.1063/5.0074631] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/31/2021] [Indexed: 05/08/2023]
Abstract
Modern neuroscience increasingly relies on 3D models to study neural circuitry, nerve regeneration, and neural disease. Several different biofabrication approaches have been explored to create 3D neural tissue model structures. Among them, 3D bioprinting has shown to have great potential to emerge as a high-throughput/high precision biofabrication strategy that can address the growing need for 3D neural models. Here, we have reviewed the design principles for neural tissue engineering. The main challenge to adapt printing technologies for biofabrication of neural tissue models is the development of neural bioink, i.e., a biomaterial with printability and gelation properties and also suitable for neural tissue culture. This review shines light on a vast range of biomaterials as well as the fundamentals of 3D neural tissue printing. Also, advances in 3D bioprinting technologies are reviewed especially for bioprinted neural models. Finally, the techniques used to evaluate the fabricated 2D and 3D neural models are discussed and compared in terms of feasibility and functionality.
Collapse
Affiliation(s)
| | - Hamed Tahmooressi
- Department of Mechanical Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Hojatollah Rezaei Nejad
- Department of Electrical and Computer Engineering, Tufts University, 161 College Avenue, Medford, Massachusetts 02155, USA
| | | | - Su-Royn Shin
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02139, USA
- Authors to whom correspondence should be addressed: and
| | - Mina Hoorfar
- Faculty of Engineering, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
- Authors to whom correspondence should be addressed: and
| |
Collapse
|