1
|
Jayakumar J, Ramesh JJ, Folkerth RD, Latha S, Kumar H, Suresh S, Srinivasan C, Kumutha J, Verma R, Sivaprakasam M. Insights into holoprosencephaly using multimodal high-resolution imaging and 3D histology. J Neuropathol Exp Neurol 2025:nlaf057. [PMID: 40411743 DOI: 10.1093/jnen/nlaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2025] Open
Abstract
Holoprosencephaly (HPE) is a well-described forebrain patterning disorder in mid-late gestation fetuses and infants. Here, we used a novel, whole-brain multimodal approach (ultrasonography, magnetic resonance imaging, and histology with 3-dimensional [3D] reconstructions with cell mapping) in earlier-gestation specimens than previously reported. In one 13- and two 22-gestational week fetuses and age-matched controls, we elucidated heretofore underappreciated HPE findings of (1) abnormal clustering of immature (doublecortin-immunoreactive) cells in the midline and paramedian forebrain, (2) linear arrays of cells in the intermediate zone of the cerebral mantle (reminiscent of subcortical band heterotopia, but possibly transient), (3) "reactive"-appearing glial fibrillary acidic protein-immunoreactive cortical cells, and (4) apparent "midline fusion" of rostral ganglionic eminences. We observed disorganization of orbitofrontal cortices and midline structures, rostral subarachnoid (marginal zone) heterotopia, and lateral displacement of the hippocampal formations utilizing multiscale multimodal 3D analytics. These findings shed light on the temporal evolution of HPE at earlier gestational ages. Moreover, this approach is scalable to include the wide range of phenotypes of HPE and is applicable to other neurologic disorders, including developmental as well as adult vascular, infectious, neoplastic, and degenerative conditions for which spatial analyses permit a fuller understanding of their pathologic spectrum.
Collapse
Affiliation(s)
- Jaikishan Jayakumar
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Center for Computational Brain Research, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Jivitha Jyothi Ramesh
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Rebecca D Folkerth
- Brain Injury Research Center, Department of Rehabilitation and Human Potential, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - S Latha
- MediScan Systems, Chennai, Tamil Nadu, India
| | | | - S Suresh
- MediScan Systems, Chennai, Tamil Nadu, India
| | - Chitra Srinivasan
- Department of Pathology, Saveetha Medical College, Chennai, Tamil Nadu, India
| | - Jayaraman Kumutha
- Department of Neonatology, Saveetha Medical College, Chennai, Tamil Nadu, India
| | - Richa Verma
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Mohanasankar Sivaprakasam
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Department of Electrical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
2
|
Patra AH, Bonefas K, Markenscoff-Papadimitriou E. "Teaching the biology of neurodiversity for social change". Dev Biol 2025:S0012-1606(25)00139-3. [PMID: 40383290 DOI: 10.1016/j.ydbio.2025.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/22/2025] [Accepted: 05/16/2025] [Indexed: 05/20/2025]
Abstract
The scientific understanding of autism spectrum disorder (ASD) is historically rooted in the diagnosis of children with divergent behavior and socialization within Western societies. Increased activism and visibility of ASD individuals has helped popularize the neurodiversity movement, which proposes that autism falls within the natural spectrum of human behavior and is not always pathological. The increased interest in neurodiversity has coincided with rapid advances in the field of psychiatric genetics, particularly in understanding the underlying genetic causes of ASD. Identified genes linked to ASD have highlighted mid-fetal cortical development as a nexus when many of these genes are co-expressed. New discoveries in the genetics of ASD present a unique opportunity in teaching developmental biology and especially brain development to engage students with the biology underlying ASD and neurodiversity in general. Informed and inclusive language, engagement with the neurodiversity community, and awareness of the complexity of the issues are good practices in teaching the biology of neurodiversity.
Collapse
Affiliation(s)
- Aindri H Patra
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY
| | - Katherine Bonefas
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY
| | | |
Collapse
|
3
|
Garcia-Martínez T, Gornatti DG, Ortiz M, Cañellas G, Heine-Suñer D, Vives-Bauzà C. The Triad of Blood-Brain Barrier Integrity: Endothelial Cells, Astrocytes, and Pericytes in Perinatal Stroke Pathophysiology. Int J Mol Sci 2025; 26:1886. [PMID: 40076511 PMCID: PMC11900453 DOI: 10.3390/ijms26051886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pediatric stroke, a significant cause of long-term neurological deficits in children, often arises from disruptions within neurovascular unit (NVU) components. The NVU, a dynamic ensemble of astrocytes, endothelial cells, pericytes, and microglia, is vital for maintaining cerebral homeostasis and regulating vascular brain development. Its structural integrity, particularly at the blood-brain barrier (BBB), depends on intercellular junctions and the basement membrane, which together restrict paracellular transport and shield the brain from systemic insults. Dysfunction in this intricate system is increasingly linked to pediatric stroke and related cerebrovascular conditions. Mutations disrupting endothelial cell adhesion or pericyte-endothelial interactions can compromise BBB stability, leading to pathological outcomes such as intraventricular hemorrhage in the germinal matrix, a hallmark of vascular brain immaturity. Additionally, inflammation, ferroptosis, necroptosis, and autophagy are key cellular processes influencing brain damage and repair. Excessive activation of these mechanisms can exacerbate NVU injury, whereas targeted therapeutic modulation offers potential pathways to mitigate damage and support recovery. This review explores the cellular and molecular mechanisms underlying NVU dysfunction, BBB disruption, and subsequent brain injury in pediatric stroke. Understanding the interplay between genetic mutations, environmental stressors, and NVU dynamics provides new insights into stroke pathogenesis. The susceptibility of the germinal matrix to vascular rupture further emphasizes the critical role of NVU integrity in early brain development. Targeting inflammatory pathways and cell death mechanisms presents promising strategies to preserve NVU function and improve outcomes for affected neonates.
Collapse
Affiliation(s)
- Tania Garcia-Martínez
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Denise G. Gornatti
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Marina Ortiz
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Guillem Cañellas
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), 07122 Palma, Spain
| | - Damià Heine-Suñer
- Genomics of Health Research, Unit of Molecular Diagnostics and Clinical Genetics, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07013 Palma, Spain;
| | - Cristòfol Vives-Bauzà
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), 07122 Palma, Spain
| |
Collapse
|
4
|
Sobierajski E, Czubay K, Beemelmans C, Beemelmans C, Meschkat M, Uhlenkamp D, Meyer G, Wahle P. Vascular Development of Fetal and Postnatal Neocortex of the Pig, the European Wild Boar Sus scrofa. J Comp Neurol 2024; 532:e70011. [PMID: 39660539 PMCID: PMC11632654 DOI: 10.1002/cne.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 12/12/2024]
Abstract
The development of the brain's vascular system is a predominantly prenatal process in mammalian species and is required for neurogenesis and further brain development. Our recent work on fetal pig has revealed that many neurodevelopmental processes start well before birth and proceed rapidly reaching near-mature status already around birth. Here, we analyzed the development of neocortical vasculature from embryonic day (E) 45 onward (gestation in pig lasts 114 days) using qualitative and quantitative image analyses and protein blots. In all cortical layers, vessel volume from total brain volume at E100 resembled that of a postnatal day (P) 30 piglet. Endothelial cells expressed the tight junction protein claudin-5 from E45 onward. GFAP+ and AQP4+ astrocytes, PDGFRβ+ pericytes, and α-SMA+ smooth muscle cells are detectable near vessels at E60 suggesting an early assembly of blood-brain barrier components. The vascular system in the visual cortex is advanced before birth with an almost mature pattern at E100. Findings were confirmed by blots that showed a steady increase of expression of tight junction and angiogenesis-related proteins (claudin-5, occludin, VE-cadherin, PECAM-1/CD31) from E65 onward until P90. The expression profile was similar in visual and somatosensory cortex. Together, we report a rapid maturation of the vascular system in pig cortex.
Collapse
Affiliation(s)
- Eric Sobierajski
- Department of Developmental Neurobiology, Faculty of Biology and BiotechnologyRuhr University BochumBochumGermany
| | - Katrin Czubay
- Department of Developmental Neurobiology, Faculty of Biology and BiotechnologyRuhr University BochumBochumGermany
| | | | | | | | | | - Gundela Meyer
- Department of Basic Medical Science, Faculty of MedicineUniversity of La LagunaSanta Cruz de TenerifeTenerifeSpain
| | - Petra Wahle
- Department of Developmental Neurobiology, Faculty of Biology and BiotechnologyRuhr University BochumBochumGermany
| |
Collapse
|
5
|
Russo ML, Sousa AMM, Bhattacharyya A. Consequences of trisomy 21 for brain development in Down syndrome. Nat Rev Neurosci 2024; 25:740-755. [PMID: 39379691 PMCID: PMC11834940 DOI: 10.1038/s41583-024-00866-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/10/2024]
Abstract
The appearance of cognitive deficits and altered brain morphology in newborns with Down syndrome (DS) suggests that these features are driven by disruptions at the earliest stages of brain development. Despite its high prevalence and extensively characterized cognitive phenotypes, relatively little is known about the cellular and molecular mechanisms that drive the changes seen in DS. Recent technical advances, such as single-cell omics and the development of induced pluripotent stem cell (iPSC) models of DS, now enable in-depth analyses of the biochemical and molecular drivers of altered brain development in DS. Here, we review the current state of knowledge on brain development in DS, focusing primarily on data from human post-mortem brain tissue. We explore the biological mechanisms that have been proposed to lead to intellectual disability in DS, assess the extent to which data from studies using iPSC models supports these hypotheses, and identify current gaps in the field.
Collapse
Affiliation(s)
- Matthew L Russo
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - André M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
6
|
Pandurangan K, Jayakumar J, Savoia S, Nanda R, Lata S, Kumar EH, S S, Vasudevan S, Srinivasan C, Joseph J, Sivaprakasam M, Verma R. Systematic development of immunohistochemistry protocol for large cryosections-specific to non-perfused fetal brain. J Neurosci Methods 2024; 405:110085. [PMID: 38387804 DOI: 10.1016/j.jneumeth.2024.110085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Immunohistochemistry (IHC) is an important technique in understanding the expression of neurochemical molecules in the developing human brain. Despite its routine application in the research and clinical setup, the IHC protocol specific for soft fragile fetal brains that are fixed using the non-perfusion method is still limited in studying the whole brain. NEW METHOD This study shows that the IHC protocols, using a chromogenic detection system, used in animals and adult humans are not optimal in the fetal brains. We have optimized key steps from Antigen retrieval (AR) to chromogen visualization for formalin-fixed whole-brain cryosections (20 µm) mounted on glass slides. RESULTS We show the results from six validated, commonly used antibodies to study the fetal brain. We achieved optimal antigen retrieval with 0.1 M Boric Acid, pH 9.0 at 70°C for 20 minutes. We also present the optimal incubation duration and temperature for protein blocking and the primary antibody that results in specific antigen labeling with minimal tissue damage. COMPARISON WITH EXISTING METHODS The IHC protocol commonly used for adult human and animal brains results in significant tissue damage in the fetal brains with little or suboptimal antigen expression. Our new method with important modifications including the temperature, duration, and choice of the alkaline buffer for AR addresses these pitfalls and provides high-quality results. CONCLUSION The optimized IHC protocol for the developing human brain (13-22 GW) provides a high-quality, repeatable, and reliable method for studying chemoarchitecture in neurotypical and pathological conditions across different gestational ages.
Collapse
Affiliation(s)
- Karthika Pandurangan
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
| | - Jaikishan Jayakumar
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India; Center for Computational Brain Research, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
| | | | - Reetuparna Nanda
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
| | - S Lata
- Mediscan Systems, Chennai, Tamil Nadu, India.
| | | | - Suresh S
- Mediscan Systems, Chennai, Tamil Nadu, India.
| | - Sudha Vasudevan
- Department of Obstetrics & Gynaecology, Saveetha Medical College, Thandalam, Chennai, Tamil Nadu, India.
| | - Chitra Srinivasan
- Department of Pathology, Saveetha Medical College, Thandalam, Chennai, Tamil Nadu, India.
| | - Jayaraj Joseph
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India; Healthcare Technology Innovation Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India; Department of Electrical Engineering, Indian Institute of Technology, Madras, Chennai, Tamil Nadu, India.
| | - Mohanasankar Sivaprakasam
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India; Healthcare Technology Innovation Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India; Department of Electrical Engineering, Indian Institute of Technology, Madras, Chennai, Tamil Nadu, India.
| | - Richa Verma
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
| |
Collapse
|
7
|
Cibelli A, Mola MG, Saracino E, Barile B, Abbrescia P, Mogni G, Spray DC, Scemes E, Rossi A, Spennato D, Svelto M, Frigeri A, Benfenati V, Nicchia GP. Aquaporin-4 and transient receptor potential vanilloid 4 balance in early postnatal neurodevelopment. Glia 2024; 72:938-959. [PMID: 38362923 DOI: 10.1002/glia.24512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/17/2024]
Abstract
In the adult brain, the water channel aquaporin-4 (AQP4) is expressed in astrocyte endfoot, in supramolecular assemblies, called "Orthogonal Arrays of Particles" (OAPs) together with the transient receptor potential vanilloid 4 (TRPV4), finely regulating the cell volume. The present study aimed at investigating the contribution of AQP4 and TRPV4 to CNS early postnatal development using WT and AQP4 KO brain and retina and neuronal stem cells (NSCs), as an in vitro model of astrocyte differentiation. Western blot analysis showed that, differently from AQP4 and the glial cell markers, TRPV4 was downregulated during CNS development and NSC differentiation. Blue native/SDS-PAGE revealed that AQP4 progressively organized into OAPs throughout the entire differentiation process. Fluorescence quenching assay indicated that the speed of cell volume changes was time-related to NSC differentiation and functional to their migratory ability. Calcium imaging showed that the amplitude of TRPV4 Ca2+ transient is lower, and the dynamics are changed during differentiation and suppressed in AQP4 KO NSCs. Overall, these findings suggest that early postnatal neurodevelopment is subjected to temporally modulated water and Ca2+ dynamics likely to be those sustaining the biochemical and physiological mechanisms responsible for astrocyte differentiation during brain and retinal development.
Collapse
Affiliation(s)
- Antonio Cibelli
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Maria Grazia Mola
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Emanuela Saracino
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Bologna, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Pasqua Abbrescia
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro-Medical School, Bari, Italy
| | - Guido Mogni
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - David C Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, NY Medical College, Valhalla, New York, USA
| | - Andrea Rossi
- Genome Engineering and Model Development Lab (GEMD), IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Diletta Spennato
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Bologna, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Frigeri
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro-Medical School, Bari, Italy
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Valentina Benfenati
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Bologna, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Bologna, Italy
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
8
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
9
|
Mulc D, Smilović D, Krsnik Ž, Junaković-Munjas A, Kopić J, Kostović I, Šimić G, Vukšić M. Fetal development of the human amygdala. J Comp Neurol 2024; 532:e25580. [PMID: 38289194 DOI: 10.1002/cne.25580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 11/03/2023] [Accepted: 12/31/2023] [Indexed: 02/01/2024]
Abstract
The intricate development of the human amygdala involves a complex interplay of diverse processes, varying in speed and duration. In humans, transient cytoarchitectural structures deliquesce, leading to the formation of functionally distinct nuclei as a result of multiple interdependent developmental events. This study compares the amygdala's cytoarchitectural development in conjunction with specific antibody reactivity for neuronal, glial, neuropil, and radial glial fibers, synaptic, extracellular matrix, and myelin components in 39 fetal human brains. We recognized that the early fetal period, as a continuation of the embryonic period, is still dominated by relatively uniform histogenetic processes. The typical appearance of ovoid cell clusters in the lateral nucleus during midfetal period is most likely associated with the cell migration and axonal growth processes in the developing human brain. Notably, synaptic markers are firstly detected in the corticomedial group of nuclei, while immunoreactivity for the panaxonal neurofilament marker SMI 312 is found dorsally. The late fetal period is characterized by a protracted migration process evidenced by the presence of doublecortin and SOX-2 immunoreactivity ventrally, in the prospective paralaminar nucleus, reinforced by vimentin immunoreactivity in the last remaining radial glial fibers. Nearing the term period, SMI 99 immunoreactivity indicates that perinatal myelination becomes prominent primarily along major axonal pathways, laying the foundation for more pronounced functional maturation. This study comprehensively elucidates the rate and sequence of maturational events in the amygdala, highlighting the key role of prenatal development in its behavioral, autonomic, and endocrine regulation, with subsequent implications for both normal functioning and psychiatric disorders.
Collapse
Affiliation(s)
- Damir Mulc
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
- Psychiatric Hospital Vrapče, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dinko Smilović
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Alisa Junaković-Munjas
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Janja Kopić
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Goran Šimić
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Mario Vukšić
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
10
|
Degl’Innocenti E, Dell’Anno MT. Human and mouse cortical astrocytes: a comparative view from development to morphological and functional characterization. Front Neuroanat 2023; 17:1130729. [PMID: 37139179 PMCID: PMC10150887 DOI: 10.3389/fnana.2023.1130729] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
The vision of astroglia as a bare scaffold to neuronal circuitry has been largely overturned. Astrocytes exert a neurotrophic function, but also take active part in supporting synaptic transmission and in calibrating blood circulation. Many aspects of their functioning have been unveiled from studies conducted in murine models, however evidence is showing many differences between mouse and human astrocytes starting from their development and encompassing morphological, transcriptomic and physiological variations when they achieve complete maturation. The evolutionary race toward superior cognitive abilities unique to humans has drastically impacted neocortex structure and, together with neuronal circuitry, astrocytes have also been affected with the acquisition of species-specific properties. In this review, we summarize diversities between murine and human astroglia, with a specific focus on neocortex, in a panoramic view that starts with their developmental origin to include all structural and molecular differences that mark the uniqueness of human astrocytes.
Collapse
Affiliation(s)
- Elisa Degl’Innocenti
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maria Teresa Dell’Anno
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- *Correspondence: Maria Teresa Dell’Anno,
| |
Collapse
|
11
|
Aragón-González A, Shaw PJ, Ferraiuolo L. Blood-Brain Barrier Disruption and Its Involvement in Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2022; 23:ijms232315271. [PMID: 36499600 PMCID: PMC9737531 DOI: 10.3390/ijms232315271] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) is a highly specialized and dynamic compartment which regulates the uptake of molecules and solutes from the blood. The relevance of the maintenance of a healthy BBB underpinning disease prevention as well as the main pathomechanisms affecting BBB function will be detailed in this review. Barrier disruption is a common aspect in both neurodegenerative diseases, such as amyotrophic lateral sclerosis, and neurodevelopmental diseases, including autism spectrum disorders. Throughout this review, conditions altering the BBB during the earliest and latest stages of life will be discussed, revealing common factors involved. Due to the barrier's role in protecting the brain from exogenous components and xenobiotics, drug delivery across the BBB is challenging. Potential therapies based on the BBB properties as molecular Trojan horses, among others, will be reviewed, as well as innovative treatments such as stem cell therapies. Additionally, due to the microbiome influence on the normal function of the brain, microflora modulation strategies will be discussed. Finally, future research directions are highlighted to address the current gaps in the literature, emphasizing the idea that common therapies for both neurodevelopmental and neurodegenerative pathologies exist.
Collapse
Affiliation(s)
- Ana Aragón-González
- Sheffield Institute for Translational Neuroscience, University of Sheffield, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK
- Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK
- Correspondence: ; Tel.: +44-(0)114-222-2257; Fax: +44-(0)114-222-2290
| |
Collapse
|
12
|
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
|
13
|
Li J, Pan L, Pembroke WG, Rexach JE, Godoy MI, Condro MC, Alvarado AG, Harteni M, Chen YW, Stiles L, Chen AY, Wanner IB, Yang X, Goldman SA, Geschwind DH, Kornblum HI, Zhang Y. Conservation and divergence of vulnerability and responses to stressors between human and mouse astrocytes. Nat Commun 2021; 12:3958. [PMID: 34172753 PMCID: PMC8233314 DOI: 10.1038/s41467-021-24232-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
Astrocytes play important roles in neurological disorders such as stroke, injury, and neurodegeneration. Most knowledge on astrocyte biology is based on studies of mouse models and the similarities and differences between human and mouse astrocytes are insufficiently characterized, presenting a barrier in translational research. Based on analyses of acutely purified astrocytes, serum-free cultures of primary astrocytes, and xenografted chimeric mice, we find extensive conservation in astrocytic gene expression between human and mouse samples. However, the genes involved in defense response and metabolism show species-specific differences. Human astrocytes exhibit greater susceptibility to oxidative stress than mouse astrocytes, due to differences in mitochondrial physiology and detoxification pathways. In addition, we find that mouse but not human astrocytes activate a molecular program for neural repair under hypoxia, whereas human but not mouse astrocytes activate the antigen presentation pathway under inflammatory conditions. Here, we show species-dependent properties of astrocytes, which can be informative for improving translation from mouse models to humans.
Collapse
Affiliation(s)
- Jiwen Li
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Lin Pan
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - William G Pembroke
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Jessica E Rexach
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Marlesa I Godoy
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Michael C Condro
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Alvaro G Alvarado
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Mineli Harteni
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Yen-Wei Chen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Endocrinology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Angela Y Chen
- Department of Obstetrics and Gynecology, University of California, Los Angeles, CA, USA
| | - Ina B Wanner
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
- Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences at UCLA, Los Angeles, CA, USA
- Brain Research Institute at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute at UCLA, Los Angeles, CA, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Daniel H Geschwind
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Harley I Kornblum
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
- Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute at UCLA, Los Angeles, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.
- Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, CA, USA.
- Brain Research Institute at UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute at UCLA, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Gupta A, Osadchiy V, Mayer EA. Brain-gut-microbiome interactions in obesity and food addiction. Nat Rev Gastroenterol Hepatol 2020; 17:655-672. [PMID: 32855515 PMCID: PMC7841622 DOI: 10.1038/s41575-020-0341-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Abstract
Normal eating behaviour is coordinated by the tightly regulated balance between intestinal and extra-intestinal homeostatic and hedonic mechanisms. By contrast, food addiction is a complex, maladaptive eating behaviour that reflects alterations in brain-gut-microbiome (BGM) interactions and a shift of this balance towards hedonic mechanisms. Each component of the BGM axis has been implicated in the development of food addiction, with both brain to gut and gut to brain signalling playing a role. Early-life influences can prime the infant gut microbiome and brain for food addiction, which might be further reinforced by increased antibiotic usage and dietary patterns throughout adulthood. The ubiquitous availability and marketing of inexpensive, highly palatable and calorie-dense food can further shift this balance towards hedonic eating through both central (disruptions in dopaminergic signalling) and intestinal (vagal afferent function, metabolic endotoxaemia, systemic immune activation, changes to gut microbiome and metabolome) mechanisms. In this Review, we propose a systems biology model of BGM interactions, which incorporates published reports on food addiction, and provides novel insights into treatment targets aimed at each level of the BGM axis.
Collapse
Affiliation(s)
- Arpana Gupta
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity Program, University of California Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Vadim Osadchiy
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity Program, University of California Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Emeran A Mayer
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity Program, University of California Los Angeles, Los Angeles, CA, USA.
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA.
- Ahmanson-Lovelace Brain Mapping Center at University of California Los Angeles, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Fleiss B, Gressens P, Stolp HB. Cortical Gray Matter Injury in Encephalopathy of Prematurity: Link to Neurodevelopmental Disorders. Front Neurol 2020; 11:575. [PMID: 32765390 PMCID: PMC7381224 DOI: 10.3389/fneur.2020.00575] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Preterm-born infants frequently suffer from an array of neurological damage, collectively termed encephalopathy of prematurity (EoP). They also have an increased risk of presenting with a neurodevelopmental disorder (e.g., autism spectrum disorder; attention deficit hyperactivity disorder) later in life. It is hypothesized that it is the gray matter injury to the cortex, in addition to white matter injury, in EoP that is responsible for the altered behavior and cognition in these individuals. However, although it is established that gray matter injury occurs in infants following preterm birth, the exact nature of these changes is not fully elucidated. Here we will review the current state of knowledge in this field, amalgamating data from both clinical and preclinical studies. This will be placed in the context of normal processes of developmental biology and the known pathophysiology of neurodevelopmental disorders. Novel diagnostic and therapeutic tactics required integration of this information so that in the future we can combine mechanism-based approaches with patient stratification to ensure the most efficacious and cost-effective clinical practice.
Collapse
Affiliation(s)
- Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Université de Paris, NeuroDiderot, Inserm, Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Helen B. Stolp
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
16
|
Jones DT, Bandopadhayay P, Jabado N. The Power of Human Cancer Genetics as Revealed by Low-Grade Gliomas. Annu Rev Genet 2019; 53:483-503. [DOI: 10.1146/annurev-genet-120417-031642] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The human brain contains a vast number of cells and shows extraordinary cellular diversity to facilitate the many cognitive and automatic commands governing our bodily functions. This complexity arises partly from large-scale structural variations in the genome, evolutionary processes to increase brain size, function, and cognition. Not surprisingly given recent technical advances, low-grade gliomas (LGGs), which arise from the glia (the most abundant cell type in the brain), have undergone a recent revolution in their classification and therapy, especially in the pediatric setting. Next-generation sequencing has uncovered previously unappreciated diverse LGG entities, unraveling genetic subgroups and multiple molecular alterations and altered pathways, including many amenable to therapeutic targeting. In this article we review these novel entities, in which oncogenic processes show striking age-related neuroanatomical specificity (highlighting their close interplay with development); the opportunities they provide for targeted therapies, some of which are already practiced at the bedside; and the challenges of implementing molecular pathology in the clinic.
Collapse
Affiliation(s)
- David T.W. Jones
- Pediatric Glioma Research Group, Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts 02215, USA
- The Broad Institute of MIT and Harvard, Boston, Massachusetts 02142, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nada Jabado
- Departments of Pediatric and Human Genetics, McGill University and the Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
17
|
Abbink MR, van Deijk ALF, Heine VM, Verheijen MH, Korosi A. The involvement of astrocytes in early-life adversity induced programming of the brain. Glia 2019; 67:1637-1653. [PMID: 31038797 PMCID: PMC6767561 DOI: 10.1002/glia.23625] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022]
Abstract
Early‐life adversity (ELA) in the form of stress, inflammation, or malnutrition, can increase the risk of developing psychopathology or cognitive problems in adulthood. The neurobiological substrates underlying this process remain unclear. While neuronal dysfunction and microglial contribution have been studied in this context, only recently the role of astrocytes in early‐life programming of the brain has been appreciated. Astrocytes serve many basic roles for brain functioning (e.g., synaptogenesis, glutamate recycling), and are unique in their capacity of sensing and integrating environmental signals, as they are the first cells to encounter signals from the blood, including hormonal changes (e.g., glucocorticoids), immune signals, and nutritional information. Integration of these signals is especially important during early development, and therefore we propose that astrocytes contribute to ELA induced changes in the brain by sensing and integrating environmental signals and by modulating neuronal development and function. Studies in rodents have already shown that ELA can impact astrocytes on the short and long term, however, a critical review of these results is currently lacking. Here, we will discuss the developmental trajectory of astrocytes, their ability to integrate stress, immune, and nutritional signals from the early environment, and we will review how different types of early adversity impact astrocytes.
Collapse
Affiliation(s)
- Maralinde R Abbink
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Lieke F van Deijk
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Vivi M Heine
- Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Mark H Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Bioengineering an Artificial Human Blood⁻Brain Barrier in Rodents. Bioengineering (Basel) 2019; 6:bioengineering6020038. [PMID: 31052208 PMCID: PMC6630638 DOI: 10.3390/bioengineering6020038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022] Open
Abstract
Our group has recently created a novel in-vivo human brain organoid vascularized with human iPSC-derived endothelial cells. In this review article, we discuss the challenges of creating a perfused human brain organoid model in an immunosuppressed rodent host and discuss potential applications for neurosurgical disease modeling.
Collapse
|
19
|
Saili KS, Zurlinden TJ, Schwab AJ, Silvin A, Baker NC, Hunter ES, Ginhoux F, Knudsen TB. Blood-brain barrier development: Systems modeling and predictive toxicology. Birth Defects Res 2018; 109:1680-1710. [PMID: 29251840 DOI: 10.1002/bdr2.1180] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/12/2017] [Indexed: 01/17/2023]
Abstract
The blood-brain barrier (BBB) serves as a gateway for passage of drugs, chemicals, nutrients, metabolites, and hormones between vascular and neural compartments in the brain. Here, we review BBB development with regard to the microphysiology of the neurovascular unit (NVU) and the impact of BBB disruption on brain development. Our focus is on modeling these complex systems. Extant in silico models are available as tools to predict the probability of drug/chemical passage across the BBB; in vitro platforms for high-throughput screening and high-content imaging provide novel data streams for profiling chemical-biological interactions; and engineered human cell-based microphysiological systems provide empirical models with which to investigate the dynamics of NVU function. Computational models are needed that bring together kinetic and dynamic aspects of NVU function across gestation and under various physiological and toxicological scenarios. This integration will inform adverse outcome pathways to reduce uncertainty in translating in vitro data and in silico models for use in risk assessments that aim to protect neurodevelopmental health.
Collapse
Affiliation(s)
- Katerine S Saili
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Todd J Zurlinden
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Andrew J Schwab
- National Health and Environmental Effects Research Laboratory (NHEERL), U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Nancy C Baker
- Leidos, contractor to NCCT, Research Triangle Park, North Carolina 27711
| | - E Sidney Hunter
- National Health and Environmental Effects Research Laboratory (NHEERL), U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Thomas B Knudsen
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| |
Collapse
|
20
|
Santoro A, Spinelli CC, Martucciello S, Nori SL, Capunzo M, Puca AA, Ciaglia E. Innate immunity and cellular senescence: The good and the bad in the developmental and aged brain. J Leukoc Biol 2018; 103:509-524. [PMID: 29389023 DOI: 10.1002/jlb.3mr0118-003r] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/21/2022] Open
Abstract
Ongoing studies evidence cellular senescence in undifferentiated and specialized cells from tissues of all ages. Although it is believed that senescence plays a wider role in several stress responses in the mature age, its participation in certain physiological and pathological processes throughout life is coming to light. The "senescence machinery" has been observed in all brain cell populations, including components of innate immunity (e.g., microglia and astrocytes). As the beneficial versus detrimental implications of senescence is an open question, we aimed to analyze the contribution of immune responses in regulatory mechanisms governing its distinct functions in healthy (development, organogenesis, danger patrolling events) and diseased brain (glioma, neuroinflammation, neurodeneration), and the putative connection between cellular and molecular events governing the 2 states. Particularly this review offers new insights into the complex roles of senescence both as a chronological event as age advances, and as a molecular mechanism of brain homeostasis through the important contribution of innate immune responses and their crosstalk with neighboring cells in brain parenchyma. We also highlight the impact of the recently described glymphatic system and brain lymphatic vasculature in the interplay between peripheral and central immune surveillance and its potential implication during aging. This will open new ways to understand brain development, its deterioration during aging, and the occurrence of several oncological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Antonietta Santoro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | | | | | - Stefania Lucia Nori
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | - Annibale Alessandro Puca
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy.,Cardiovascular Research Unit, IRCCS MultiMedica, Milan, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| |
Collapse
|
21
|
Lin MY, Wang YL, Wu WL, Wolseley V, Tsai MT, Radic V, Thornton ME, Grubbs BH, Chow RH, Huang IC. Zika Virus Infects Intermediate Progenitor Cells and Post-mitotic Committed Neurons in Human Fetal Brain Tissues. Sci Rep 2017; 7:14883. [PMID: 29093521 PMCID: PMC5665882 DOI: 10.1038/s41598-017-13980-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/05/2017] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) infection is associated with microcephaly in fetuses, but the pathogenesis of ZIKV-related microcephaly is not well understood. Here we show that ZIKV infects the subventricular zone in human fetal brain tissues and that the tissue tropism broadens with the progression of gestation. Our research demonstrates also that intermediate progenitor cells (IPCs) are the main target cells for ZIKV. Post-mitotic committed neurons become susceptible to ZIKV infection as well at later stages of gestation. Furthermore, activation of microglial cells, DNA fragmentation, and apoptosis of infected or uninfected cells could be found in ZIKV-infected brain tissues. Our studies identify IPCs as the main target cells for ZIKV. They also suggest that immune activation after ZIKV infection may play an important role in the pathogenesis of ZIKV-related microcephaly.
Collapse
Affiliation(s)
- Ming-Yi Lin
- Department of Physiology & Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yi-Ling Wang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Wan-Lin Wu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Victoria Wolseley
- Department of Physiology & Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ming-Ting Tsai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Vladimir Radic
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew E Thornton
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brendan H Grubbs
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert H Chow
- Department of Physiology & Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - I-Chueh Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Mottahedin A, Ardalan M, Chumak T, Riebe I, Ek J, Mallard C. Effect of Neuroinflammation on Synaptic Organization and Function in the Developing Brain: Implications for Neurodevelopmental and Neurodegenerative Disorders. Front Cell Neurosci 2017; 11:190. [PMID: 28744200 PMCID: PMC5504097 DOI: 10.3389/fncel.2017.00190] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/20/2017] [Indexed: 12/27/2022] Open
Abstract
The brain is a plastic organ where both the intrinsic CNS milieu and extrinsic cues play important roles in shaping and wiring neural connections. The perinatal period constitutes a critical time in central nervous system development with extensive refinement of neural connections, which are highly sensitive to fetal and neonatal compromise, such as inflammatory challenges. Emerging evidence suggests that inflammatory cells in the brain such as microglia and astrocytes are pivotal in regulating synaptic structure and function. In this article, we will review the role of glia cells in synaptic physiology and pathophysiology, including microglia-mediated elimination of synapses. We propose that activation of the immune system dynamically affects synaptic organization and function in the developing brain. We will discuss the role of neuroinflammation in altered synaptic plasticity following perinatal inflammatory challenges and potential implications for neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Amin Mottahedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Tetyana Chumak
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Ilse Riebe
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| |
Collapse
|
23
|
Mishra A. Binaural blood flow control by astrocytes: listening to synapses and the vasculature. J Physiol 2016; 595:1885-1902. [PMID: 27619153 DOI: 10.1113/jp270979] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/15/2016] [Indexed: 12/28/2022] Open
Abstract
Astrocytes are the most common glial cells in the brain with fine processes and endfeet that intimately contact both neuronal synapses and the cerebral vasculature. They play an important role in mediating neurovascular coupling (NVC) via several astrocytic Ca2+ -dependent signalling pathways such as K+ release through BK channels, and the production and release of arachidonic acid metabolites. They are also involved in maintaining the resting tone of the cerebral vessels by releasing ATP and COX-1 derivatives. Evidence also supports a role for astrocytes in maintaining blood pressure-dependent change in cerebrovascular tone, and perhaps also in blood vessel-to-neuron signalling as posited by the 'hemo-neural hypothesis'. Thus, astrocytes are emerging as new stars in preserving the intricate balance between the high energy demand of active neurons and the supply of oxygen and nutrients from the blood by maintaining both resting blood flow and activity-evoked changes therein. Following neuropathology, astrocytes become reactive and many of their key signalling mechanisms are altered, including those involved in NVC. Furthermore, as they can respond to changes in vascular pressure, cardiovascular diseases might exert previously unknown effects on the central nervous system by altering astrocyte function. This review discusses the role of astrocytes in neurovascular signalling in both physiology and pathology, and the impact of these findings on understanding BOLD-fMRI signals.
Collapse
Affiliation(s)
- Anusha Mishra
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
24
|
Krencik R, van Asperen JV, Ullian EM. Human astrocytes are distinct contributors to the complexity of synaptic function. Brain Res Bull 2016; 129:66-73. [PMID: 27570101 DOI: 10.1016/j.brainresbull.2016.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/07/2016] [Accepted: 08/22/2016] [Indexed: 01/03/2023]
Abstract
Cellular components of synaptic circuits have been adjusted for increased human brain size, neural cell density, energy consumption and developmental duration. How does the human brain make these accommodations? There is evidence that astrocytes are one of the most divergent neural cell types in primate brain evolution and it is now becoming clear that they have critical roles in controlling synaptic development, function and plasticity. Yet, we still do not know how the precise developmental appearance of these cells and subsequent astrocyte-derived signals modulate diverse neuronal circuit subtypes. Here, we discuss what is currently known about the influence of glial factors on synaptic maturation and focus on unique features of human astrocytes including their potential roles in regenerative and translational medicine. Human astrocyte distinctiveness may be a major contributor to high level neuronal processing of the human brain and act in novel ways during various neuropathies ranging from autism spectrum disorders, viral infection, injury and neurodegenerative conditions.
Collapse
Affiliation(s)
- Robert Krencik
- Departments of Ophthalmology and Physiology, Neuroscience Program, University of California San Francisco, United States.
| | - Jessy V van Asperen
- Departments of Ophthalmology and Physiology, Neuroscience Program, University of California San Francisco, United States
| | - Erik M Ullian
- Departments of Ophthalmology and Physiology, Neuroscience Program, University of California San Francisco, United States
| |
Collapse
|
25
|
Silbereis JC, Pochareddy S, Zhu Y, Li M, Sestan N. The Cellular and Molecular Landscapes of the Developing Human Central Nervous System. Neuron 2016; 89:248-68. [PMID: 26796689 DOI: 10.1016/j.neuron.2015.12.008] [Citation(s) in RCA: 504] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The human CNS follows a pattern of development typical of all mammals, but certain neurodevelopmental features are highly derived. Building the human CNS requires the precise orchestration and coordination of myriad molecular and cellular processes across a staggering array of cell types and over a long period of time. Dysregulation of these processes affects the structure and function of the CNS and can lead to neurological or psychiatric disorders. Recent technological advances and increased focus on human neurodevelopment have enabled a more comprehensive characterization of the human CNS and its development in both health and disease. The aim of this review is to highlight recent advancements in our understanding of the molecular and cellular landscapes of the developing human CNS, with focus on the cerebral neocortex, and the insights these findings provide into human neural evolution, function, and dysfunction.
Collapse
Affiliation(s)
- John C Silbereis
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sirisha Pochareddy
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ying Zhu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mingfeng Li
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics and Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
26
|
Rao VT, Ludwin SK, Fuh SC, Sawaya R, Moore CS, Ho MK, Bedell BJ, Sarnat HB, Bar-Or A, Antel JP. MicroRNA Expression Patterns in Human Astrocytes in Relation to Anatomical Location and Age. J Neuropathol Exp Neurol 2016; 75:156-66. [DOI: 10.1093/jnen/nlv016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
27
|
Baker SJ, Ellison DW, Gutmann DH. Pediatric gliomas as neurodevelopmental disorders. Glia 2015; 64:879-95. [PMID: 26638183 DOI: 10.1002/glia.22945] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/13/2015] [Indexed: 01/01/2023]
Abstract
Brain tumors represent the most common solid tumor of childhood, with gliomas comprising the largest fraction of these cancers. Several features distinguish them from their adult counterparts, including their natural history, causative genetic mutations, and brain locations. These unique properties suggest that the cellular and molecular etiologies that underlie their development and maintenance might be different from those that govern adult gliomagenesis and growth. In this review, we discuss the genetic basis for pediatric low-grade and high-grade glioma in the context of developmental neurobiology, and highlight the differences between histologically-similar tumors arising in children and adults.
Collapse
Affiliation(s)
- Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude's Children's Research Hospital, Memphis, Tennessee
| | - David W Ellison
- Department of Pathology, St. Jude's Children's Research Hospital, Memphis, Tennessee
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
28
|
Rottkamp DM, Rudenko IA, Maier MT, Roshanbin S, Yulyaningsih E, Perez L, Valdearcos M, Chua S, Koliwad SK, Xu AW. Leptin potentiates astrogenesis in the developing hypothalamus. Mol Metab 2015; 4:881-9. [PMID: 26629411 PMCID: PMC4632125 DOI: 10.1016/j.molmet.2015.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 08/18/2015] [Accepted: 08/24/2015] [Indexed: 12/05/2022] Open
Abstract
Background The proper establishment of hypothalamic feeding circuits during early development has a profound influence on energy homeostasis, and perturbing this process could predispose individuals to obesity and its associated consequences later in life. The maturation of hypothalamic neuronal circuitry in rodents takes place during the initial postnatal weeks, and this coincides with a dramatic surge in the circulating level of leptin, which is known to regulate the outgrowth of key neuronal projections in the maturing hypothalamus. Coincidently, this early postnatal period also marks the rapid proliferation and expansion of astrocytes in the brain. Methods Here we examined the effects of leptin on the proliferative capacity of astrocytes in the developing hypothalamus by treating postnatal mice with leptin. Mutant mice were also generated to conditionally remove leptin receptors from glial fibrillary acidic protein (GFAP)-expressing cells in the postnatal period. Results and conclusions We show that GFAP-expressing cells in the periventricular zone of the 3rd ventricle were responsive to leptin during the initial postnatal week. Leptin enhanced the proliferation of astrocytes in the postnatal hypothalamus and conditional removal of leptin receptors from GFAP-expressing cells during early postnatal period limited astrocyte proliferation. While increasing evidence demonstrates a direct role of leptin in regulating astrocytes in the adult brain, and given the essential function of astrocytes in modulating neuronal function and connectivity, our study indicates that leptin may exert its metabolic effects, in part, by promoting hypothalamic astrogenesis during early postnatal development. GFAP-expressing cells in the periventricular zone of 3rd ventricle are leptin-responsive during the initial postnatal week. Leptin enhances the proliferation of astrocytes in the postnatal hypothalamus. Conditional removal of leptin receptors in GFAP-expressing cells in early postnatal period reduces astrocyte proliferation.
Collapse
Affiliation(s)
- Daniele M Rottkamp
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ivan A Rudenko
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew T Maier
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sahar Roshanbin
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ernie Yulyaningsih
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Luz Perez
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Streamson Chua
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Suneil K Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Allison W Xu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
29
|
Remyelination After Cuprizone-Induced Demyelination Is Accelerated in Juvenile Mice. J Neuropathol Exp Neurol 2015; 74:756-66. [DOI: 10.1097/nen.0000000000000214] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
30
|
Fern R. Ischemic tolerance in pre-myelinated white matter: the role of astrocyte glycogen in brain pathology. J Cereb Blood Flow Metab 2015; 35:951-8. [PMID: 25669910 PMCID: PMC4640254 DOI: 10.1038/jcbfm.2015.3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/15/2014] [Accepted: 12/17/2014] [Indexed: 11/09/2022]
Abstract
In isolated white matter, ischemic tolerance changes dramatically in the period immediately before the onset of myelination. In the absence of an extrinsic energy source, postnatal day 0 to 2 (P0 to P2) white matter axons are here shown to maintain excitability for over twice as long as axons >P2, a differential that was dependent on glycogen metabolism. Prolonged withdrawal of extrinsic energy supply tended to spare axons in zones around astrocytes, which are shown to be the sole repository for glycogen particles in developing white matter. Analysis of mitochondrial volume fraction revealed that neither axons nor astrocytes had a low metabolic rate in neonatal white matter, while oligodendroglia at older ages had an elevated metabolism. The astrocyte population is established early in neural development, and exhibits reduced cell density as maturation progresses and white matter expands. The findings show that this event establishes the necessary conditions for ischemia sensitivity in white matter and indicates that astrocyte proximity may be significant for the survival of neuronal elements in conditions associated with compromised energy supply.
Collapse
Affiliation(s)
- Robert Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| |
Collapse
|
31
|
Huria T, Beeraka NM, Al-Ghamdi B, Fern R. Premyelinated central axons express neurotoxic NMDA receptors: relevance to early developing white-matter injury. J Cereb Blood Flow Metab 2015; 35:543-53. [PMID: 25515212 PMCID: PMC4420873 DOI: 10.1038/jcbfm.2014.227] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/12/2014] [Accepted: 11/19/2014] [Indexed: 01/05/2023]
Abstract
Ischemic-type injury to developing white matter is associated with the significant clinical condition cerebral palsy and with the cognitive deficits associated with premature birth. Premyelinated axons are the major cellular component of fetal white matter and loss of axon function underlies the disability, but the cellular mechanisms producing ischemic injury to premyelinated axons have not previously been described. Injury was found to require longer periods of modelled ischemia than at latter developmental points. Ischemia produced initial hyperexcitability in axons followed by loss of function after Na(+) and Ca(2+) influx. N-methyl-D-aspartate- (NMDA) type glutamate receptor (GluR) agonists potentiated axon injury while antagonists were protective. The NMDA GluR obligatory Nr1 subunit colocalized with markers of small premyelinated axons and expression was found at focal regions of axon injury. Ischemic injury of glial cells present in early developing white matter was NMDA GluR independent. Axons in human postconception week 18 to 23 white matter had a uniform prediameter expansion phenotype and postembedded immuno-gold labelling showed Nr1 subunit expression on the membrane of these axons, demonstrating a shared key neuropathologic feature with the rodent model. Premyelinated central axons therefore express high levels of functional NMDA GluRs that confer sensitivity to ischemic injury.
Collapse
Affiliation(s)
- Tahani Huria
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | | | - Badrah Al-Ghamdi
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | - Robert Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, John Bull Building, Research Way, Plymouth, UK
| |
Collapse
|
32
|
Hansen DB, Garrido-Comas N, Salter M, Fern R. HCO3(-)-independent pH regulation in astrocytes in situ is dominated by V-ATPase. J Biol Chem 2015; 290:8039-47. [PMID: 25666621 PMCID: PMC4375461 DOI: 10.1074/jbc.m115.636597] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The mechanisms of HCO3−-independent intracellular pH (pHi) regulation were examined in fibrous astrocytes within isolated neonatal rat optic nerve (RON) and in cultured cortical astrocytes. In agreement with previous studies, resting pHi in cultured astrocytes was 6.82 ± 0.06 and inhibition of the V-ATPase H+ pump by Cl− removal or via the selective inhibitor bafilomycin had only a small effect upon resting pHi and recovery following an acid load. In contrast, resting pHi in RON astrocytes was 7.10 ± 0.04, significantly less acidic than that in cultured cells (p < 0.001), and responded to inhibition of V-ATPase with profound acidification to the 6.3–6.5 range. Fluorescent immuno-staining and immuno-gold labeling confirmed the presence V-ATPase in the cell membrane of RON astrocyte processes and somata. Using ammonia pulse recovery, pHi recovery in RON astrocyte was achieved largely via V-ATPase with sodium-proton exchange (NHE) playing a minor role. The findings indicate that astrocytes in a whole-mount preparation such as the optic nerve rely to a greater degree upon V-ATPase for HCO3−-independent pHi regulation than do cultured astrocytes, with important functional consequences for the regulation of pH in the CNS.
Collapse
Affiliation(s)
- Daniel Bloch Hansen
- From the Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU
| | - Nestor Garrido-Comas
- the Department of Cell Physiology and Pharmacology, University of Leicester, Leicester LE1 7RH, and
| | - Mike Salter
- the Institute of Membrane and Systems Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Robert Fern
- From the Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU,
| |
Collapse
|
33
|
Verkhratsky A, Nedergaard M, Hertz L. Why are astrocytes important? Neurochem Res 2014; 40:389-401. [PMID: 25113122 DOI: 10.1007/s11064-014-1403-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/22/2014] [Accepted: 07/26/2014] [Indexed: 12/27/2022]
Abstract
Astrocytes, which populate the grey and white mater of the brain and the spinal cord are highly heterogeneous in their morphology and function. These cells are primarily responsible for homeostasis of the central nervous system (CNS). Most central synapses are surrounded by exceedingly thin astroglial perisynaptic processes, which act as "astroglial cradle" critical for genesis, maturation and maintenance of synaptic connectivity. The perisynaptic glial processes are densely packed with numerous transporters, which provide for homeostasis of ions and neurotransmitters in the synaptic cleft, for local metabolic support and for release of astroglial derived scavengers of reactive oxygen species. Through perivascular processes astrocytes contribute to blood-brain barrier and form "glymphatic" drainage system of the CNS. Furthermore astrocytes are indispensible for glutamatergic and γ-aminobutyrate-ergic synaptic transmission being the supplier of neurotransmitters precursor glutamine via an astrocytic/neuronal cycle. Pathogenesis of many neurological disorders, including neuropsychiatric and neurodegenerative diseases is defined by loss of homeostatic function (astroglial asthenia) or remodelling of astroglial homoeostatic capabilities. Astroglial cells further contribute to neuropathologies through mounting complex defensive programme generally known as reactive astrogliosis.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK,
| | | | | |
Collapse
|
34
|
Fern RF, Matute C, Stys PK. White matter injury: Ischemic and nonischemic. Glia 2014; 62:1780-9. [PMID: 25043122 DOI: 10.1002/glia.22722] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Ischemic pathologies of white matter (WM) include a large proportion of stroke and developmental lesions while multiple sclerosis (MS) is the archetype nonischemic pathology. Growing evidence suggests other important diseases including neurodegenerative and psychiatric disorders also involve a significant WM component. Axonal, oligodendroglial, and astroglial damage proceed via distinct mechanisms in ischemic WM and these mechanisms evolve dramatically with maturation. Axons may pass through four developmental stages where the pattern of membrane protein expression influences how the structure responds to ischemia; WM astrocytes pass through at least two and differ significantly in their ischemia tolerance from grey matter astrocytes; oligodendroglia pass through at least three, with the highly ischemia intolerant pre-oligodendrocyte (pre-Oli) stage linking the less sensitive precursor and mature phenotypes. Neurotransmitters play a central role in WM pathology at all ages. Glutamate excitotoxicity in WM has both necrotic and apoptotic components; the latter mediated by intracellular pathways which differ between receptor types. ATP excitotoxicity may be largely mediated by the P2X7 receptor and also has both necrotic and apoptotic components. Interplay between microglia and other cell types is a critical element in the injury process. A growing appreciation of the significance of WM injury for nonischemic neurological disorders is currently stimulating research into mechanisms; with curious similarities being found with those operating during ischemia. A good example is traumatic brain injury, where axonal pathology can proceed via almost identical pathways to those described during acute ischemia.
Collapse
Affiliation(s)
- Robert F Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, United Kingdom
| | | | | |
Collapse
|
35
|
Atik A, Cheong J, Harding R, Rees S, De Matteo R, Tolcos M. Impact of daily high-dose caffeine exposure on developing white matter of the immature ovine brain. Pediatr Res 2014; 76:54-63. [PMID: 24739937 DOI: 10.1038/pr.2014.55] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/16/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Caffeine is widely used to treat apnea of prematurity, but the standard dosing regimen is not always sufficient to prevent apnea. Before higher doses of caffeine can be used, their effects on the immature brain need to be carefully evaluated. Our aim was to determine the impact of daily high-dose caffeine administration on the developing white matter of the immature ovine brain. METHODS High-dose caffeine (25 mg/kg caffeine base loading dose; 20 mg/kg daily maintenance dose; n = 9) or saline (n = 8) were administered to pregnant sheep from 0.7 to 0.8 of term, equivalent to approximately 27-34 wk in humans. At 0.8 of term, the white and gray matter were assessed histologically and immunohistochemically. RESULTS Daily caffeine administration led to peak caffeine concentration of 32 mg/l in fetal plasma at 1 h, followed by a gradual decline, with no effects on mean arterial pressure and heart rate. Initial caffeine exposure led to transient, mild alkalosis in the fetus but did not alter oxygenation. At necropsy, there was no effect of daily high-dose caffeine on brain weight, oligodendrocyte density, myelination, axonal integrity, microgliosis, astrogliosis, apoptosis, or neuronal density. CONCLUSION Daily high-dose caffeine administration does not appear to adversely affect the developing white matter at the microstructural level.
Collapse
Affiliation(s)
- Anzari Atik
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Jeanie Cheong
- 1] Department of Neonatal Services, Royal Women's Hospital, Victorian Infant Brain Studies, Murdoch Children's Research Institute, University of Melbourne, Melbourne, Victoria, Australia [2] Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Sandra Rees
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Robert De Matteo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Mary Tolcos
- The Ritchie Centre, MIMR-PHI Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| |
Collapse
|
36
|
Borre YE, O'Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med 2014; 20:509-18. [PMID: 24956966 DOI: 10.1016/j.molmed.2014.05.002] [Citation(s) in RCA: 750] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 02/06/2023]
Abstract
Gut microbiota is essential to human health, playing a major role in the bidirectional communication between the gastrointestinal tract and the central nervous system. The microbiota undergoes a vigorous process of development throughout the lifespan and establishes its symbiotic rapport with the host early in life. Early life perturbations of the developing gut microbiota can impact neurodevelopment and potentially lead to adverse mental health outcomes later in life. This review compares the parallel early development of the intestinal microbiota and the nervous system. The concept of parallel and interacting microbial-neural critical windows opens new avenues for developing novel microbiota-modulating based therapeutic interventions in early life to combat neurodevelopmental deficits and brain disorders.
Collapse
Affiliation(s)
- Yuliya E Borre
- Laboratory of NeuroGastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital, Cork, Ireland
| | - Gerard Clarke
- Laboratory of NeuroGastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Department of Psychiatry, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Timothy G Dinan
- Laboratory of NeuroGastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F Cryan
- Laboratory of NeuroGastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
37
|
Hepatitis C virus (HCV) interaction with astrocytes: nonproductive infection and induction of IL-18. J Neurovirol 2014; 20:278-93. [PMID: 24671718 DOI: 10.1007/s13365-014-0245-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/10/2014] [Accepted: 02/20/2014] [Indexed: 12/15/2022]
Abstract
Hepatitis C virus (HCV) infection causes the central nervous system (CNS) abnormalities in more than 50 % of chronically infected subjects. However, the underlying mechanisms are largely unknown. In this study, we characterized the HCV interactions with astrocytes, one of the putative HCV target cells in the brain. We demonstrated that primary human astrocytes (PHA) were very inefficiently infected by HCV, either in the cell-free form or through cell-cell contact. We then determined the potential restriction steps of HCV infection and replication in these cells. PHA expressed all known HCV receptors but failed to support HCV entry. HCV IRES-mediated RNA translation was functional in PHA and further enhanced by miR122 expression. Nevertheless, PHA did not support HCV replication regardless of miR122 expression. To our great surprise, we found that HCV exposure induced robust IL-18 expression in PHA and exhibited direct neurotoxicity. Taken together, these results showed that astrocytes did not support productive HCV infection and replication, but HCV interactions with astrocytes and neurons alone might be sufficient to cause CNS dysfunction.
Collapse
|
38
|
Semple BD, Blomgren K, Gimlin K, Ferriero DM, Noble-Haeusslein LJ. Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol 2013; 106-107:1-16. [PMID: 23583307 PMCID: PMC3737272 DOI: 10.1016/j.pneurobio.2013.04.001] [Citation(s) in RCA: 1487] [Impact Index Per Article: 123.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/03/2013] [Accepted: 04/03/2013] [Indexed: 12/13/2022]
Abstract
Hypoxic-ischemic and traumatic brain injuries are leading causes of long-term mortality and disability in infants and children. Although several preclinical models using rodents of different ages have been developed, species differences in the timing of key brain maturation events can render comparisons of vulnerability and regenerative capacities difficult to interpret. Traditional models of developmental brain injury have utilized rodents at postnatal day 7-10 as being roughly equivalent to a term human infant, based historically on the measurement of post-mortem brain weights during the 1970s. Here we will examine fundamental brain development processes that occur in both rodents and humans, to delineate a comparable time course of postnatal brain development across species. We consider the timing of neurogenesis, synaptogenesis, gliogenesis, oligodendrocyte maturation and age-dependent behaviors that coincide with developmentally regulated molecular and biochemical changes. In general, while the time scale is considerably different, the sequence of key events in brain maturation is largely consistent between humans and rodents. Further, there are distinct parallels in regional vulnerability as well as functional consequences in response to brain injuries. With a focus on developmental hypoxic-ischemic encephalopathy and traumatic brain injury, this review offers guidelines for researchers when considering the most appropriate rodent age for the developmental stage or process of interest to approximate human brain development.
Collapse
Affiliation(s)
- Bridgette D. Semple
- Department of Neurological Surgery, University of California San Francisco, 513 Parnassus Avenue, Room HSE-722, San Francisco, CA 94143-0112, USA
| | - Klas Blomgren
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
- Department of Pediatrics, Queen Silvia's Children's Hospital, University of Gothenburg, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Q2:07, SE 171 76 Stockholm, Sweden
| | - Kayleen Gimlin
- Department of Neurological Surgery, University of California San Francisco, 513 Parnassus Avenue, Room HSE-722, San Francisco, CA 94143-0112, USA
| | - Donna M. Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Linda J. Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, 513 Parnassus Avenue, Room HSE-722, San Francisco, CA 94143-0112, USA
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
39
|
Rusu MC, Dermengiu D, Loreto C, Motoc AGM, Pop E. Astrocitary niches in human adult medulla oblongata. Acta Histochem 2013; 115:296-300. [PMID: 22909904 DOI: 10.1016/j.acthis.2012.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 07/15/2012] [Accepted: 07/16/2012] [Indexed: 11/24/2022]
Abstract
Astrocytes are considered as neuromodulators of the CNS. Whereas experimental studies on astrocitary functions are gaining importance, the anatomy of the astrocitary niches in the human CNS has been overlooked. The study was performed on the brainstem of 10 adult cadavers. We aimed to determine astrocitary niches in the human medulla oblongata using immunohistochemical labeling with vimentin and also CD34 immunostaining to accurately diagnose associated microvessels. Niches rich in astrocytes were identified as follows: (a) the superficial layer of astrocytes, ventral and ventrolateral, in the rostral medulla oblongata; (b) the median raphe; (c) medullary nuclei: arcuate nucleus, area postrema, nucleus of the solitary tract; (d) the subependymal zone (SEZ, caudal medulla) and subventricular zone (SVZ, rostral medulla). Astrocytes were scarce in the ventrolateral medulla, and mostly present within the pyramidal tract and the olivary nucleus. Apart from the SEZ and SVZ, the brainstem niches of astrocytes mostly overlap those regions known to perform roles as central respiratory chemoreceptors. The astrocytes of the SEZ and SVZ, which are known as stem cell niches, are related to an increased microvascular density.
Collapse
|
40
|
Alix JJP, Zammit C, Riddle A, Meshul CK, Back SA, Valentino M, Fern R. Central axons preparing to myelinate are highly sensitive [corrected] to ischemic injury. Ann Neurol 2013; 72:936-51. [PMID: 23280842 DOI: 10.1002/ana.23690] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/22/2012] [Accepted: 06/15/2012] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Developing central white matter is subject to ischemic-type injury during the period that precedes myelination. At this stage in maturation, central axons initiate a program of radial expansion and ion channel redistribution. Here we test the hypothesis that during radial expansion axons display heightened ischemic sensitivity, when clusters of Ca(2+) channels decorate future node of Ranvier sites. METHODS Functionality and morphology of central axons and glia were examined during and after a period of modeled ischemia. Pathological changes in axons undergoing radial expansion were probed using electrophysiological, quantitative ultrastructural, and morphometric analysis in neonatal rodent optic nerve and periventricular white matter axons studied under modeled ischemia in vitro or after hypoxia-ischemia in vivo. RESULTS Acute ischemic injury of central axons undergoing initial radial expansion was mediated by Ca(2+) influx through Ca(2+) channels expressed in axolemma clusters. This form of injury operated only in this axon population, which was more sensitive to injury than neighboring myelinated axons, smaller axons yet to initiate radial expansion, astrocytes, or oligodendroglia. A pharmacological strategy designed to protect both small and large diameter premyelinated axons proved 100% protective against acute ischemia studied under modeled ischemia in vitro or after hypoxia-ischemia in vivo. INTERPRETATION Recent clinical data highlight the importance of axon pathology in developing white matter injury. The elevated susceptibility of early maturing axons to ischemic injury described here may significantly contribute to selective white matter pathology and places these axons alongside preoligodendrocytes as a potential primary target of both injury and therapeutics.
Collapse
Affiliation(s)
- James J P Alix
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
41
|
Capper D, Mittelbronn M, Goeppert B, Meyermann R, Schittenhelm J. Secreted protein, acidic and rich in cysteine (SPARC) expression in astrocytic tumour cells negatively correlates with proliferation, while vascular SPARC expression is associated with patient survival. Neuropathol Appl Neurobiol 2010; 36:183-97. [DOI: 10.1111/j.1365-2990.2010.01072.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Trivedi R, Gupta RK, Husain N, Rathore RKS, Saksena S, Srivastava S, Malik GK, Das V, Pradhan M, Sarma MK, Pandey CM, Narayana PA. Region-specific maturation of cerebral cortex in human fetal brain: diffusion tensor imaging and histology. Neuroradiology 2009; 51:567-76. [PMID: 19421746 DOI: 10.1007/s00234-009-0533-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 04/22/2009] [Indexed: 11/28/2022]
Abstract
INTRODUCTION In this study, diffusion tensor imaging (DTI) and glial fibrillary acidic protein (GFAP) immunohistochemical analysis in different cortical regions in fetal brains at different gestational age (GA) were performed. METHODS DTI was performed on 50 freshly aborted fetal brains with GA ranging from 12 to 42 weeks to compare age-related fractional anisotropy (FA) changes in different cerebral cortical regions that include frontal, parietal, occipital, and temporal lobes at the level of thalami. GFAP immunostaining was performed and the percentage of GFAP-positive areas was quantified. RESULTS The cortical FA values in the frontal lobe peaked at around 26 weeks of GA, occipital and temporal lobes at around 20 weeks, and parietal lobe at around 23 weeks. A significant, but modest, positive correlation (r = 0.31, p = 0.02) was observed between cortical FA values and percentage area of GFAP expression in cortical region around the time period during which the migrational events are at its peak, i.e., GA < or = 28 weeks for frontal cortical region and GA < or = 22 weeks for rest of the lobes. CONCLUSIONS The DTI-derived FA quantification with its GFAP immunohistologic correlation in cortical regions of the various lobes of the cerebral hemispheres supports region-specific migrational and maturational events in human fetal brain.
Collapse
Affiliation(s)
- Richa Trivedi
- Department of Radiodiagnosis, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, UP, India, 226014
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dieni S, Inder T, Yoder B, Briscoe T, Camm E, Egan G, Denton D, Rees S. The Pattern of Cerebral Injury in a Primate Model of Preterm Birth and Neonatal Intensive Care. J Neuropathol Exp Neurol 2004; 63:1297-309. [PMID: 15624766 DOI: 10.1093/jnen/63.12.1297] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Survivors of very premature birth face an increased risk of adverse motor, cognitive, and behavior sequelae. In order to understand the pathogenesis of these adverse outcomes, an animal model of premature birth and neonatal care in a species with a close similarity to the human infant is sought. In this histological and immunohistochemical study we have defined the pattern of cerebral injury in a premature baboon model undergoing similar neonatal intensive care to that of the human premature infant. Sixteen baboons were delivered at 125 days gestation (dg; term approximately184 dg) with 14 days neonatal intensive care and were compared with gestational control brains at 125, 140, and 160 dg. The premature baboons undergoing neonatal intensive care sustained a spectrum of neuropathologies including white matter injury, hemorrhage, and ventriculomegaly, which resemble lesions frequently observed in the human premature infant. These data suggest that the premature baboon is a model with similarities in maturation and pattern of cerebral injury to the human infant that may provide useful insights of relevance to the human preterm infant.
Collapse
Affiliation(s)
- Sandra Dieni
- Department of Anatomy and Cell Biology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Kálmán M. Glial reaction and reactive glia. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
45
|
Abstract
Hypotheses are presented to explain the pathogenesis of several clinical features of holoprosencephaly, and neuropathologic approaches to testing these hypotheses are suggested. The traditional morphologic classification of holoprosencephaly into alobar, semilobar, and lobar forms is grades of severity, and each occurs in all of the genetic mutations known. Of the four defective genes identified as primary in human holoprosencephaly, three exhibit a ventrodorsal gradient of expression (SHH, SIX3, and TGIF) and one a dorsoventral gradient (ZIC2). But, in addition to the vertical axis, genes expressed in the neural tube also may have rostrocaudal and mediolateral gradients in the other axes. These other gradients may be equally as important as the vertical. If the rostrocaudal gradient extends as far as the mesencephalic neuromere, it may interfere with the formation, migration, or apoptosis of the mesencephalic neural crest, which forms membranous bones of the face, orbits, nose, and parts of the eyes, and may explain the midfacial hypoplasia seen in many, but not all, children with holoprosencephaly. This rostrocaudal gradient also causes noncleavage of the caudate nucleus, thalamus, and hypothalamus and contributes to the formation of the dorsal cyst of holoprosencephaly, which is probably derived from an expanded suprapineal recess of the 3rd ventricle with secondary dilation of the telencephalic monoventricle and at times may produce a unique transfontanellar encephalocele. The extent of the mediolateral gradient may explain the severe disorganization of cerebral cortical architecture in medial parts of the forebrain and normal cortex in lateral parts, including the radial glial fibers. This preserved lateral cortex may explain why some children with holoprosencephaly have better intellectual function than expected and may also be important in the pathogenesis of epilepsy, by contrast with malformations such as lissencephaly, in which the entire cerebral cortex is involved. Epilepsy in some, but not all, cases also may be related to the sequential maturation of axonal terminals in relation to the neurons they innervate. Diabetes insipidus is a complication in a majority of patients; other neuroendocrinopathies occur less frequently. Secondary down-regulation of the OTP gene or of downstream genes such as BRN2 or SIM1 may result in failure of terminal differentiation of magnocellular neurons of the supraoptic and paraventricular hypothalamic nuclei. Disoriented radial glial fibers or abnormal ependyma may allow aberrant migration of neuroepithelial cells into the ventricle. A new classification of holoprosencephaly is needed to integrate morphologic and genetic criteria.
Collapse
Affiliation(s)
- H B Sarnat
- Department of Pathology, University of Washington School of Medicine, Seattle, USA.
| | | |
Collapse
|
46
|
Ajtai BM, Kállai L, Kálmán M. Capability for reactive gliosis develops prenatally in the diencephalon but not in the cortex of rats. Exp Neurol 1997; 146:151-8. [PMID: 9225748 DOI: 10.1006/exnr.1997.6496] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this study, the glial reactions to stab wounds were investigated on a large population of newborn (P0) and fetal rats, by the immunohistochemical staining of the glial fibrillary acidic protein. The lesions penetrated both the cortex and the diencephalon. The fetuses were lesioned in utero from the 17th embryonic day (E17) and were born on E22 or E23 in the natural way. In the cortex usually no reactive gliosis developed although definitive tissue destructions remained after the lesion. Weak and incomplete glial reactions were observed in a few cases of E20 or P0 lesions only. In the diencephalon, however, the same stabbings provoked massive glial reactions. The timing and the morphology of this reaction were similar to those found in adult animals. At E17 the lesion did not result in reactive gliosis even in the diencephalon. Our study highlights two phenomena: (i) depending on the brain area servere glial reactions can already follow fetal lesions, and (ii) the appearance of the capability for glial reactions may be a stage of the local tissue maturation in every brain area and cannot be considered as a function of brain development in general. Probably, the capability for glial reactions can take place only when certain histogenetic processes (e.g., cell migration, axon growth, apoptosis) have been at least mostly accomplished, but which of the local development events are the determining ones remains to be investigated.
Collapse
Affiliation(s)
- B M Ajtai
- Department of Anatomy, Histology and Embryology, Semmelweis University of Medicine, Budapest, Hungary
| | | | | |
Collapse
|
47
|
Blennow M, Rosengren L, Jonsson S, Forssberg H, Katz-Salamon M, Hagberg H, Hesser U, Lagercrantz H. Glial fibrillary acidic protein is increased in the cerebrospinal fluid of preterm infants with abnormal neurological findings. Acta Paediatr 1996; 85:485-9. [PMID: 8740311 DOI: 10.1111/j.1651-2227.1996.tb14068.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Glial fibrillary acidic protein (GFAP) is the structural protein of the intermediate filament of astroglia. The aims of the present study were to examine GFAP in the cerebrospinal fluid (CSF) of preterm infants at different postmenstrual ages and to evaluate the potential of GFAP to predict abnormal neurodevelopmental outcome. GFAP increased in correlation with postmenstrual age in preterm infants (n = 17) and full-term infants (n = 9). The levels were five times higher in preterm infants (n = 10) with an abnormal neonatal course and/or an abnormal neurological outcome than in healthy preterm infants. The positive predictive value of a GFAP higher than the 98th percentile of normal infants was 69%, while a GFAP level below this limit invariable predicted a good outcome. Simultaneously analysed noradrenaline, hypoxanthine and glutamate did not differ between the groups. We conclude that CSF GFAP increases with maturity and that CSF GFAP appears to be a promising marker for perinatal brain damage.
Collapse
Affiliation(s)
- M Blennow
- Department of Woman and Child Health, Karolinska Institute Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Pundt LL, Kondoh T, Conrad JA, Low WC. Transplantation of human striatal tissue into a rodent model of Huntington's disease: phenotypic expression of transplanted neurons and host-to-graft innervation. Brain Res Bull 1996; 39:23-32. [PMID: 8846104 DOI: 10.1016/0361-9230(95)02029-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The present study was undertaken to investigate the phenotypic expression and integration of human striatal neurons transplanted into an animal model of Huntington's disease. Sprague-Dawley rats were anesthetized and subjected to quinolinic acid lesions of the left striatum. Three human fetal cadavers were utilized for transplantation in this study (7, 8, and 10 weeks in gestation). The striatal primordia was dissected from each fetus and subsequently dissociated into cell suspensions. Following the initial lesion surgeries (3-4 months), the rats were reanesthetized and transplanted with human striatal cells (400,000 cells per rat). The animals were processed for histochemical analysis 9-17 weeks posttransplantation. Histochemistry was performed utilizing thionin (Nissl staining), acetylcholinesterase, NADPH-diaphorase, and antibodies against tyrosine hydroxylase and glial fibrillary acidic protein. Examination of stained brain sections demonstrate that human striatal transplants grow to fill a substantial portion of the remaining striatum, and contain clusters of immature and mature cells. Acetylcholinesterase activity is present in the transplant neuropil, varying in intensity, and distributed in a heterogeneous fashion. In addition, host afferent dopaminergic fibers penetrate into the transplant, and are occasionally found in patches. NADPH-diaphorase histochemistry revealed medium sized aspiny striatal neurons of donor origin in the transplants. The results of this study are similar to those obtained with rodent fetal striatal transplants, and suggest that human striatal tissue is capable of surviving, expressing normal striatal cell phenotypes, and receiving host dopaminergic innervation.
Collapse
Affiliation(s)
- L L Pundt
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
49
|
Pundt LL, Kondoh T, Low WC. The fate of human glial cells following transplantation in normal rodents and rodent models of neurodegenerative disease. Brain Res 1995; 695:25-36. [PMID: 8574644 DOI: 10.1016/0006-8993(95)00753-d] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Investigations on xenografting in the brain have previously focused on the anatomical and functional integration of the transplanted neurons. More recently, astrocytes are being implicated as having complex functions following transplantation, and are being investigated to determine their role(s) in transplantation. The present study was undertaken to investigate the migration of human astrocytes following transplantation of thalamic, striatal, and mesencephalic tissue into the rodent striatum. Human donor fetuses (9-16 weeks in gestation) obtained through elective and spontaneous abortions were utilized in this study. Following transplantation, donor astrocytes were labeled with an antiserum directed against human glial fibrillary acidic protein. Our results demonstrate that astrocytic elements from all three tissue types are capable of incorporating into the host brain, and have a tendency to follow white matter tracts (such as the corpus callosum, internal capsule, and fiber bundles in the striatum). Human astrocytes, originating from the striatum and thalamus exhibited extensive migration, while migration was more limited in animals with ventral mesencephalon transplants. Ventral mesencephalon transplanted animal demonstrated positive astrocytes within the transplant, with processes (very few cell bodies) extending into white matter of adjacent host striatum. Astrocytes demonstrating immature morphology were observed with all transplant types, but were most prevalent in the striatal transplanted animals. The extent of astrocyte migration and the morphologies observed in this study reflect regional differences of the developing human brain. These results confirm and extend previous investigations on glial cell migration following transplantation in the brain.
Collapse
Affiliation(s)
- L L Pundt
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis 55455, USA
| | | | | |
Collapse
|
50
|
Rivkin MJ, Flax J, Mozell R, Osathanondh R, Volpe JJ, Villa-Komaroff L. Oligodendroglial development in human fetal cerebrum. Ann Neurol 1995; 38:92-101. [PMID: 7611731 DOI: 10.1002/ana.410380116] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We have successfully established mixed glial cell primary cultures prepared from individual fetal human brains (15-18 weeks' gestation in age). Cultures were maintained for as long as 3 months in either 10% fetal calf serum (FCS) or serum-free chemically defined medium (CDM). By morphological and immunohistochemical criteria, the precursor cell for human oligodendrocytes (O-2A cell) was identified. This cell exhibited the bipolar morphology and A2B5-positive (A2B5+) immunoreactivity typical of the O-2A precursor cell. With time in culture, cells possessing a stellate morphology appeared, some of which stained with the O4 antibody, indicative of cell differentiation in the oligodendroglial lineage. At yet older culture age, arborized cells bearing the O1 (galactocerebroside, GC) immunohistochemical marker and displaying the morphological characteristics typical of more mature oligodendrocytes were found, confirming their oligodendroglial identity. Oligodendroglial differentiation was supported best by CDM rather than FCS. To complement these observations, double immunofluorescent studies were performed on parietal sections from human fetal brains at 20 to 22 weeks of gestation. Bipolar A2B5+, multipolar A2B5+/O4+, and arborized A2B5-/O1+ cells were found, thus confirming the presence of oligodendrocytes in human fetal brain at this stage of prenatal development and consistent with the observations made in cell culture.
Collapse
Affiliation(s)
- M J Rivkin
- Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|