1
|
Sarnat HB, Yu W. Keratan sulfate proteoglycan: putative template for neuroblast migratory and axonal fascicular pathways and fetal expression in globus pallidus, thalamus, and olfactory bulb. J Neuropathol Exp Neurol 2025; 84:8-21. [PMID: 38950418 DOI: 10.1093/jnen/nlae057] [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] [Indexed: 07/03/2024] Open
Abstract
Keratan sulfate (KS) is a proteoglycan secreted in the fetal brain astrocytes and radial glia into extracellular parenchyma as granulofilamentous deposits. KS surrounds neurons except dendritic spines, repelling glutamatergic and facilitating GABAergic axons. The same genes are expressed in both neuroblast migration and axonal growth. This study examines timing of KS during morphogenesis of some normally developing human fetal forebrain structures. Twenty normal human fetal brains from 9-41 weeks gestational age were studied at autopsy. KS was examined by immunoreactivity in formalin-fixed paraffin sections, plus other markers including synaptophysin, S-100β protein, vimentin and nestin. Radial and tangential neuroblast migratory pathways from subventricular zone to cortical plate were marked by KS deposits as early as 9wk GA, shortly after neuroblast migration initiated. During later gestation this reactivity gradually diminished and disappeared by term. Long axonal fascicles of the internal capsule and short fascicles of intrinsic bundles of globus pallidus and corpus striatum also appeared as early as 9-12wk, as fascicular sleeves before axons even entered. Intense KS occurs in astrocytic cytoplasm and extracellular parenchyma at 9wk in globus pallidus, 15wk thalamus, 18wk corpus striatum, 22wk cortical plate, and hippocampus postnatally. Corpus callosum and anterior commissure do not exhibit KS at any age. Optic chiasm shows reactivity at the periphery but not around intrinsic subfasciculi. We postulate that KS forms a chemical template for many long and short axonal fascicles before axons enter and neuroblast migratory pathways at initiation of migration. Cross-immunoreactivity with aggrecan may render difficult molecular distinction.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Neuropathology, Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Paediatrics, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Departments of Paediatrics and Pathology (Neuropathology), Owerko Centre, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Weiming Yu
- Anatomical Pathology, Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| |
Collapse
|
2
|
González-Arnay E, Pérez-Santos I, Jiménez-Sánchez L, Cid E, Gal B, de la Prida LM, Cavada C. Immunohistochemical field parcellation of the human hippocampus along its antero-posterior axis. Brain Struct Funct 2024; 229:359-385. [PMID: 38180568 PMCID: PMC10917878 DOI: 10.1007/s00429-023-02725-9] [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: 04/15/2023] [Accepted: 10/15/2023] [Indexed: 01/06/2024]
Abstract
The primate hippocampus includes the dentate gyrus, cornu ammonis (CA), and subiculum. CA is subdivided into four fields (CA1-CA3, plus CA3h/hilus of the dentate gyrus) with specific pyramidal cell morphology and connections. Work in non-human mammals has shown that hippocampal connectivity is precisely patterned both in the laminar and longitudinal axes. One of the main handicaps in the study of neuropathological semiology in the human hippocampus is the lack of clear laminar and longitudinal borders. The aim of this study was to explore a histochemical segmentation of the adult human hippocampus, integrating field (medio-lateral), laminar, and anteroposterior longitudinal patterning. We provide criteria for head-body-tail field and subfield parcellation of the human hippocampus based on immunodetection of Rabphilin3a (Rph3a), Purkinje-cell protein 4 (PCP4), Chromogranin A and Regulation of G protein signaling-14 (RGS-14). Notably, Rph3a and PCP4 allow to identify the border between CA3 and CA2, while Chromogranin A and RGS-14 give specific staining of CA2. We also provide novel histological data about the composition of human-specific regions of the anterior and posterior hippocampus. The data are given with stereotaxic coordinates along the longitudinal axis. This study provides novel insights for a detailed region-specific parcellation of the human hippocampus useful for human brain imaging and neuropathology.
Collapse
Affiliation(s)
- Emilio González-Arnay
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Basic Medical Science-Division of Human Anatomy, Universidad de La Laguna, Santa Cruz de Tenerife, Canary Islands, Spain
| | - Isabel Pérez-Santos
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lorena Jiménez-Sánchez
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Elena Cid
- Instituto Cajal, CSIC, Madrid, Spain
| | - Beatriz Gal
- Instituto Cajal, CSIC, Madrid, Spain
- Universidad CEU-San Pablo, Madrid, Spain
| | | | - Carmen Cavada
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
3
|
Sarnat HB, Flores-Sarnat L. Neuroembryonic and fetal brain development: Relevance to fetal/neonatal neurological training. Semin Fetal Neonatal Med 2024; 29:101520. [PMID: 38679531 DOI: 10.1016/j.siny.2024.101520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Insight into neuroembryology, developmental neuroanatomy and neurophysiology distinguish the diagnostic approaches of paediatric from adult neurologists and general paediatricians. These fundamental disciplines of basic neuroscience could be more effectively taught during paediatric neurology and most residency programmes, that will strengthen career-long learning. Interdisciplinary training of fetal-neonatal neurology within these programs requires working knowledge of neuroembryology applied to maternal reproductive health influencing the maternal-placental-fetal triad, neonate, and young child. Systematic didactic teaching of development in terms of basic neuroscience with neuropathological context would better address needed clinical skill sets to be incorporated into paediatric neurology and neonatology residencies to address brain health and diseases across childhood. Trainees need to recognize the continuity of development, established by maternal reproductive health before conception with gene -environment influences over the first 1000 days. Considerations of neuroembryology that explain earlier brain development during the first half of pregnancy enhances an understanding of effects throughout gestation through parturition and into neonatal life. Neonatal EEG training enhances these clinical descriptions by applying serial EEG-state analyses of premature neonates through early childhood to recognize evolving patterns associated with neuronal maturation and synaptogenesis. Neuroimaging studies offer comparisons of normal structural images with malformations and destructive lesions to correlate with clinical and neurophysiological findings. This analysis better assesses aberrant developmental processes in the context of neuroembryology. Time-specific developmental events and semantic precision are important for accurate phenotypic descriptions for a better understanding of etiopathogenesis with maturation. Certification of paediatric neurology training programme curricula should apply practical knowledge of basic neuroscience in the context of nervous system development and maturation from conception through postnatal time periods. Interdisciplinary fetal-neonatal neurology training constitutes an important educational component for career-long learning.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics (Neurology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada; Pathology and Laboratory Medicine (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada.
| | - Laura Flores-Sarnat
- Departments of Paediatrics (Neurology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
| |
Collapse
|
4
|
Shen J, Zhang T, Guan H, Li X, Zhang S, Xu G. PDGFR-beta signaling mediates endogenous neurogenesis after postischemic neural stem/progenitor cell transplantation in mice. Brain Inj 2023; 37:1345-1354. [PMID: 37975626 DOI: 10.1080/02699052.2023.2280894] [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: 04/23/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Although platelet-derived growth factor receptor (PDGFR)-β mediates the self-renewal and multipotency of neural stem/progenitor cells (NSPCs) in vitro and in vivo, its mechanisms of activating endogenous NSPCs following ischemic stroke still remain unproven. METHODS The exogenous NSPCs were transplanted into the ischemic striatum of PDGFR-β conditionally neuroepithelial knockout (KO) mice at 24 h after transient middle cerebral artery occlusion (tMCAO). 5-Bromo-2'-deoxyuridine (BrdU) was intraperitoneally injected to label the newly formed endogenous NSPCs. Infarction volume was measured, and behavioral tests were performed. In the subventricular zone (SVZ), proliferation of endogenous NSPCs was tested, and synapse formation and expression of nutritional factors were measured. RESULTS Compared with control mice, KO mice showed larger infarction volume, delayed neurological recovery, reduced numbers of BrdU positive cells, decreased expression of neurogenic factors (including neurofilament, synaptophysin, and brain-derived neurotrophic factor), and decreased synaptic regeneration in SVZ after tMCAO. Moreover, exogenous NSPC transplantation significantly alleviated neurologic dysfunction, promoted neurogenesis, increased expression of neurologic factors, and diminished synaptic deformation in SVZ of FL mice after tMCAO but had no beneficial effect in KO mice. CONCLUSION PDGFR-β signaling may promote activation of endogenous NSPCs after postischemic NSPC transplantation, and thus represents a novel potential regeneration-based therapeutic target.
Collapse
Affiliation(s)
- Jie Shen
- Department of Neurology, Dongguan Binhaiwan Central Hospital, Dongguan, Guang Dong, China
| | - Tong Zhang
- School of Medicine, Shanxi Datong University, Datong, Shanxi, China
- Institute of Brain Science, Shanxi Datong University, Datong, Shanxi, China
| | - Hong Guan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xin Li
- Department of Pulmonary and Critical Care Medicine, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Sainan Zhang
- Department of Pulmonary and Critical Care Medicine, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Guihua Xu
- Department of Science and Education, Dongguan Binhaiwan Central Hospital, Dongguan, Guang Dong, China
- Dongguan Key Laboratory of Precision Medicine
| |
Collapse
|
5
|
Sarnat HB, Chan ES, Ng D, Yu W. Maturation of metastases in peripheral neuroblastic tumors (neuroblastoma) of children. J Neuropathol Exp Neurol 2023; 82:853-864. [PMID: 37682248 DOI: 10.1093/jnen/nlad068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023] Open
Abstract
Peripheral neuroblastic tumors of childhood exhibit 3 principal neural crest lineages: primitive neuroblastoma, ganglioneuroblastoma, and ganglioneuroma. They are unique in undergoing maturation of neurons (ganglion cells) and Schwann cells, thereby recapitulating normal fetal neuronal development in the brain. Precision in estimating neurogenesis is enhanced by immunoreactivities of markers of neuronal maturation. Whether organ tissue factors in different sites of metastases influence rates of maturation and whether metastases are similar to their primary neuroblastic tumor are incompletely documented. Four young children, 1 with a mixed primary adrenal tumor and 3 with metastases were studied at surgery or autopsy. Immunocytochemical reactivities included microtubule-associated protein-2, synaptophysin, chromogranin-A, somatostatin, keratan sulfate, vimentin, S-100β protein, and PHOX2B. Primary tumors were non-uniform with regions of either poor or enhanced maturation. Both neuronal and Schwannian lineages were represented in each tumor type but differed in proportions. Bi- or multi-nucleated ganglion cells matured equal to mononuclear forms. Ganglion cell maturation was similar in metastases regardless of the target organ. Metastases resembled primary tumors. Immunocytochemical markers of neuronal and of Schwann cell maturation provide greater diagnostic precision to supplement histological criteria. Interval between diagnosis of primary tumor and metastases, metastatic target tissues, and chemotherapy over an interval of time do not appear to influence neuroblastic or Schwann cell differentiation.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Department of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
| | - Elaine S Chan
- Department of Pathology and Laboratory Medicine (Paediatric Anatomical Pathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
| | - Denise Ng
- Department of Pathology and Laboratory Medicine (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
| | - Weiming Yu
- Department of Pathology and Laboratory Medicine (Paediatric Anatomical Pathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
| |
Collapse
|
6
|
Song B, Zhang Y, Xiong G, Luo H, Zhang B, Li Y, Wang Z, Zhou Z, Chang X. Single-cell transcriptomic analysis reveals the adverse effects of cadmium on the trajectory of neuronal maturation. Cell Biol Toxicol 2023; 39:1697-1713. [PMID: 36114956 DOI: 10.1007/s10565-022-09775-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022]
Abstract
Cadmium (Cd) is an extensively existing environmental pollutant that has neurotoxic effects. However, the molecular mechanism of Cd on neuronal maturation is unveiled. Single-cell RNA sequencing (scRNA-seq) has been widely used to uncover cellular heterogeneity and is a powerful tool to reconstruct the developmental trajectory of neurons. In this study, neural stem cells (NSCs) from subventricular zone (SVZ) of newborn mice were treated with CdCl2 for 24 h and differentiated for 7 days to obtain neuronal lineage cells. Then scRNA-seq analysis identified five cell stages with different maturity in neuronal lineage cells. Our findings revealed that Cd altered the trajectory of maturation of neuronal lineage cells by decreasing the number of cells in different stages and hindering their maturation. Cd induced differential transcriptome expression in different cell subpopulations in a stage-specific manner. Specifically, Cd induced oxidative damage and changed the proportion of cell cycle phases in the early stage of neuronal development. Furthermore, the autocrine and paracrine signals of Wnt5a were downregulated in the low mature neurons in response to Cd. Importantly, activation of Wnt5a effectively rescued the number of neurons and promoted their maturation. Taken together, the findings of this study provide new and comprehensive insights into the adverse effect of Cd on neuronal maturation.
Collapse
Affiliation(s)
- Bo Song
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Yuwei Zhang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Guiya Xiong
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Huan Luo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Bing Zhang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Yixi Li
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Zhibin Wang
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Xiuli Chang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Lebedeva OS, Sharova EI, Grekhnev DA, Skorodumova LO, Kopylova IV, Vassina EM, Oshkolova A, Novikova IV, Krisanova AV, Olekhnovich EI, Vigont VA, Kaznacheyeva EV, Bogomazova AN, Lagarkova MA. An Efficient 2D Protocol for Differentiation of iPSCs into Mature Postmitotic Dopaminergic Neurons: Application for Modeling Parkinson's Disease. Int J Mol Sci 2023; 24:7297. [PMID: 37108456 PMCID: PMC10139404 DOI: 10.3390/ijms24087297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
About 15% of patients with parkinsonism have a hereditary form of Parkinson's disease (PD). Studies on the early stages of PD pathogenesis are challenging due to the lack of relevant models. The most promising ones are models based on dopaminergic neurons (DAns) differentiated from induced pluripotent stem cells (iPSCs) of patients with hereditary forms of PD. This work describes a highly efficient 2D protocol for obtaining DAns from iPSCs. The protocol is rather simple, comparable in efficiency with previously published protocols, and does not require viral vectors. The resulting neurons have a similar transcriptome profile to previously published data for neurons, and have a high level of maturity marker expression. The proportion of sensitive (SOX6+) DAns in the population calculated from the level of gene expression is higher than resistant (CALB+) DAns. Electrophysiological studies of the DAns confirmed their voltage sensitivity and showed that a mutation in the PARK8 gene is associated with enhanced store-operated calcium entry. The study of high-purity DAns differentiated from the iPSCs of patients with hereditary PD using this differentiation protocol will allow for investigators to combine various research methods, from patch clamp to omics technologies, and maximize information about cell function in normal and pathological conditions.
Collapse
Affiliation(s)
- Olga S. Lebedeva
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Elena I. Sharova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Dmitriy A. Grekhnev
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Liubov O. Skorodumova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Irina V. Kopylova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Ekaterina M. Vassina
- Vavilov Institute of General Genetics, GSP-1, Gubkina St., 3, 119991 Moscow, Russia
| | - Arina Oshkolova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Iuliia V. Novikova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Alena V. Krisanova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Evgenii I. Olekhnovich
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Vladimir A. Vigont
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Elena V. Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Alexandra N. Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Maria A. Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| |
Collapse
|
8
|
Sarnat HB. Sequences of synaptogenesis in the human fetal and neonatal brain by synaptophysin immunocytochemistry. Front Cell Neurosci 2023; 17:1105183. [PMID: 36816854 PMCID: PMC9936616 DOI: 10.3389/fncel.2023.1105183] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/02/2023] [Indexed: 02/05/2023] Open
Abstract
Synaptogenesis is the final phase of axonal pathfinding. Its sequences of spatial and temporal development in the immature nervous system are precisely timed and consistent. Synaptophysin, a principal structural glycoprotein of synaptic vesicle membranes regardless of the chemical transmitter substance within, is a reliable means of demonstrating sequences of synaptogenesis in human fetal brain tissue at autopsy and is resistant to postmortem autolysis. Furthermore, synaptophysin molecules are demonstrated during axoplasmic flow before being assembled into membranes in immature axons and also mature axons of neurons with a high metabolic rate. In brain malformations these sequences often are altered both in distribution of synapses and in timing, often delayed but sometimes precocious, with postnatal clinical manifestations such as epilepsy and cognitive development.
Collapse
Affiliation(s)
- Harvey B. Sarnat
- Department of Paediatrics (Neurology), Pathology and Laboratory Medicine (Neuropathology) and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children’s Hospital Research Institute (Owerko Centre), Calgary, AB, Canada
| |
Collapse
|
9
|
Mukhtar T, Breda J, Adam MA, Boareto M, Grobecker P, Karimaddini Z, Grison A, Eschbach K, Chandrasekhar R, Vermeul S, Okoniewski M, Pachkov M, Harwell CC, Atanasoski S, Beisel C, Iber D, van Nimwegen E, Taylor V. Temporal and sequential transcriptional dynamics define lineage shifts in corticogenesis. EMBO J 2022; 41:e111132. [PMID: 36345783 PMCID: PMC9753470 DOI: 10.15252/embj.2022111132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
The cerebral cortex contains billions of neurons, and their disorganization or misspecification leads to neurodevelopmental disorders. Understanding how the plethora of projection neuron subtypes are generated by cortical neural stem cells (NSCs) is a major challenge. Here, we focused on elucidating the transcriptional landscape of murine embryonic NSCs, basal progenitors (BPs), and newborn neurons (NBNs) throughout cortical development. We uncover dynamic shifts in transcriptional space over time and heterogeneity within each progenitor population. We identified signature hallmarks of NSC, BP, and NBN clusters and predict active transcriptional nodes and networks that contribute to neural fate specification. We find that the expression of receptors, ligands, and downstream pathway components is highly dynamic over time and throughout the lineage implying differential responsiveness to signals. Thus, we provide an expansive compendium of gene expression during cortical development that will be an invaluable resource for studying neural developmental processes and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tanzila Mukhtar
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Jeremie Breda
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Manal A Adam
- Eli and Edythe Broad Center of Regeneration Medicine and Stem cell ResearchUniversity of California, San FranciscoSan FranciscoCAUSA
- Weill Institute for NeuroscienceSan FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Marcelo Boareto
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Computational Biology Group, D‐BSSEETH ZürichBaselSwitzerland
| | - Pascal Grobecker
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Zahra Karimaddini
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Computational Biology Group, D‐BSSEETH ZürichBaselSwitzerland
| | - Alice Grison
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Katja Eschbach
- Department of Biosystems Science and EngineeringETH ZürichBaselSwitzerland
| | | | - Swen Vermeul
- Scientific IT ServicesETH ZürichZürichSwitzerland
| | | | - Mikhail Pachkov
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Corey C Harwell
- Eli and Edythe Broad Center of Regeneration Medicine and Stem cell ResearchUniversity of California, San FranciscoSan FranciscoCAUSA
- Weill Institute for NeuroscienceSan FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Suzana Atanasoski
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Faculty of MedicineUniversity of ZürichZürichSwitzerland
| | - Christian Beisel
- Department of Biosystems Science and EngineeringETH ZürichBaselSwitzerland
| | - Dagmar Iber
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Weill Institute for NeuroscienceSan FranciscoCAUSA
| | - Erik van Nimwegen
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Verdon Taylor
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| |
Collapse
|
10
|
Reddy DS, Abeygunaratne HN. Experimental and Clinical Biomarkers for Progressive Evaluation of Neuropathology and Therapeutic Interventions for Acute and Chronic Neurological Disorders. Int J Mol Sci 2022; 23:11734. [PMID: 36233034 PMCID: PMC9570151 DOI: 10.3390/ijms231911734] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/27/2022] Open
Abstract
This article describes commonly used experimental and clinical biomarkers of neuronal injury and neurodegeneration for the evaluation of neuropathology and monitoring of therapeutic interventions. Biomarkers are vital for diagnostics of brain disease and therapeutic monitoring. A biomarker can be objectively measured and evaluated as a proxy indicator for the pathophysiological process or response to therapeutic interventions. There are complex hurdles in understanding the molecular pathophysiology of neurological disorders and the ability to diagnose them at initial stages. Novel biomarkers for neurological diseases may surpass these issues, especially for early identification of disease risk. Validated biomarkers can measure the severity and progression of both acute neuronal injury and chronic neurological diseases such as epilepsy, migraine, Alzheimer's disease, Parkinson's disease, Huntington's disease, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis, and other brain diseases. Biomarkers are deployed to study progression and response to treatment, including noninvasive imaging tools for both acute and chronic brain conditions. Neuronal biomarkers are classified into four core subtypes: blood-based, immunohistochemical-based, neuroimaging-based, and electrophysiological biomarkers. Neuronal conditions have progressive stages, such as acute injury, inflammation, neurodegeneration, and neurogenesis, which can serve as indices of pathological status. Biomarkers are critical for the targeted identification of specific molecules, cells, tissues, or proteins that dramatically alter throughout the progression of brain conditions. There has been tremendous progress with biomarkers in acute conditions and chronic diseases affecting the central nervous system.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Intercollegiate School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Hasara Nethma Abeygunaratne
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
11
|
Pesce L, Scardigli M, Gavryusev V, Laurino A, Mazzamuto G, Brady N, Sancataldo G, Silvestri L, Destrieux C, Hof PR, Costantini I, Pavone FS. 3D molecular phenotyping of cleared human brain tissues with light-sheet fluorescence microscopy. Commun Biol 2022; 5:447. [PMID: 35551498 PMCID: PMC9098858 DOI: 10.1038/s42003-022-03390-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/21/2022] [Indexed: 12/17/2022] Open
Abstract
The combination of optical tissue transparency with immunofluorescence allows the molecular characterization of biological tissues in 3D. However, adult human organs are particularly challenging to become transparent because of the autofluorescence contributions of aged tissues. To meet this challenge, we optimized SHORT (SWITCH-H2O2-antigen Retrieval-TDE), a procedure based on standard histological treatments in combination with a refined clearing procedure to clear and label portions of the human brain. 3D histological characterization with multiple molecules is performed on cleared samples with a combination of multi-colors and multi-rounds labeling. By performing fast 3D imaging of the samples with a custom-made inverted light-sheet fluorescence microscope (LSFM), we reveal fine details of intact human brain slabs at subcellular resolution. Overall, we proposed a scalable and versatile technology that in combination with LSFM allows mapping the cellular and molecular architecture of the human brain, paving the way to reconstruct the entire organ.
Collapse
Affiliation(s)
- Luca Pesce
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
| | - Marina Scardigli
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
| | - Vladislav Gavryusev
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
| | - Annunziatina Laurino
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Giacomo Mazzamuto
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Italy
| | - Niamh Brady
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Giuseppe Sancataldo
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Italy
| | | | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Irene Costantini
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Italy.
- Department of Biology, University of Florence, Florence, Italy.
| | - Francesco S Pavone
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Italy
| |
Collapse
|
12
|
Alonso-Alconada D, Gressens P, Golay X, Robertson NJ. Neurogenesis Is Reduced at 48 h in the Subventricular Zone Independent of Cell Death in a Piglet Model of Perinatal Hypoxia-Ischemia. Front Pediatr 2022; 10:793189. [PMID: 35573964 PMCID: PMC9106110 DOI: 10.3389/fped.2022.793189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular and tissue damage triggered after hypoxia-ischemia (HI) can be generalized and affect the neurogenic niches present in the central nervous system. As neuroregeneration may be critical for optimizing functional recovery in neonatal encephalopathy, the goal of the present work was to investigate the neurogenic response to HI in the neurogenic niche of the subventricular zone (SVZ) in the neonatal piglet. A total of 13 large white male piglets aged <24 h were randomized into two groups: i) HI group (n = 7), animals submitted to transient cerebral HI and resuscitation; and ii) Control group (n = 6), non-HI animals. At 48 h, piglets were euthanized, and the SVZ and its surrounding regions, such as caudate and periventricular white matter, were analyzed for histology using hematoxylin-eosin staining and immunohistochemistry by evaluating the presence of cleaved caspase 3 and TUNEL positive cells, together with the cell proliferation/neurogenesis markers Ki67 (cell proliferation), GFAP (neural stem cells processes), Sox2 (neural stem/progenitor cells), and doublecortin (DCX, a marker of immature migrating neuroblasts). Hypoxic-ischemic piglets showed a decrease in cellularity in the SVZ independent of cell death, together with decreased length of neural stem cells processes, neuroblast chains area, DCX immunoreactivity, and lower number of Ki67 + and Ki67 + Sox2 + cells. These data suggest a reduction in both cell proliferation and neurogenesis in the SVZ of the neonatal piglet, which could in turn compromise the replacement of the lost neurons and the achievement of global repair.
Collapse
Affiliation(s)
- Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Xavier Golay
- Department of Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
| | - Nicola J. Robertson
- Institute for Women’s Health, University College London, London, United Kingdom
- Edinburgh Neuroscience, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Transitory and Vestigial Structures of the Developing Human Nervous System. Pediatr Neurol 2021; 123:86-101. [PMID: 34416613 DOI: 10.1016/j.pediatrneurol.2021.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 11/23/2022]
Abstract
As with many body organs, the human central nervous system contains many structures and cavities that may have had functions in embryonic and fetal life but are vestigial or atrophic at maturity. Examples are the septum pellucidum, remnants of the lamina terminalis, Cajal-Retzius neurons, induseum griseum, habenula, and accessory olfactory bulb. Other structures are transitory in fetal or early postnatal life, disappearing from the mature brain. Examples are the neural crest, subpial granular glial layer of Brun over cerebral cortex, radial glial cells, and subplate zone of cerebral cortex. At times persistent fetal structures that do not regress may cause neurological problems or indicate a pathologic condition, such as Blake pouch cyst. Transitory structures thus can become vestigial. Examples are an excessively wide cavum septi pellucidi, suprapineal recess of the third ventricle, trigeminal artery of the posterior fossa circulation, and hyaloid ocular artery. Arrested maturation might be considered another aspect of vestigial structure. An example is the persistent microcolumnar cortical architecture in focal cortical dysplasia type Ia, in cortical zones of chronic fetal ischemia, and in some metabolic/genetic congenital encephalopathies. Some transitory structures in human brain are normal adult structures in lower vertebrates. Recognition of transitory and vestigial structures by fetal or postnatal neuroimaging and neuropathologically enables better understanding of cerebral ontogenesis and avoids misinterpretations.
Collapse
|
14
|
Exploring the human cerebral cortex using confocal microscopy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 168:3-9. [PMID: 34536443 PMCID: PMC8992370 DOI: 10.1016/j.pbiomolbio.2021.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/07/2021] [Indexed: 01/12/2023]
Abstract
Cover-all mapping of the distribution of neurons in the human brain would have a significant impact on the deep understanding of brain function. Therefore, complete knowledge of the structural organization of different human brain regions at the cellular level would allow understanding their role in the functions of specific neural networks. Recent advances in tissue clearing techniques have allowed important advances towards this goal. These methods use specific chemicals capable of dissolving lipids, making the tissue completely transparent by homogenizing the refractive index. However, labeling and clearing human brain samples is still challenging. Here, we present an approach to perform the cellular mapping of the human cerebral cortex coupling immunostaining with SWITCH/TDE clearing and confocal microscopy. A specific evaluation of the contributions of the autofluorescence signals generated from the tissue fixation is provided as well as an assessment of lipofuscin pigments interference. Our evaluation demonstrates the possibility of obtaining an efficient clearing and labeling process of parts of adult human brain slices, making it an excellent method for morphological classification and antibody validation of neuronal and non-neuronal markers.
Collapse
|
15
|
Narayanasamy KK, Stojic A, Li Y, Sass S, Hesse MR, Deussner-Helfmann NS, Dietz MS, Kuner T, Klevanski M, Heilemann M. Visualizing Synaptic Multi-Protein Patterns of Neuronal Tissue With DNA-Assisted Single-Molecule Localization Microscopy. Front Synaptic Neurosci 2021; 13:671288. [PMID: 34220481 PMCID: PMC8247585 DOI: 10.3389/fnsyn.2021.671288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/25/2021] [Indexed: 02/04/2023] Open
Abstract
The development of super-resolution microscopy (SRM) has widened our understanding of biomolecular structure and function in biological materials. Imaging multiple targets within a single area would elucidate their spatial localization relative to the cell matrix and neighboring biomolecules, revealing multi-protein macromolecular structures and their functional co-dependencies. SRM methods are, however, limited to the number of suitable fluorophores that can be imaged during a single acquisition as well as the loss of antigens during antibody washing and restaining for organic dye multiplexing. We report the visualization of multiple protein targets within the pre- and postsynapse in 350–400 nm thick neuronal tissue sections using DNA-assisted single-molecule localization microscopy (SMLM). In a single labeling step, antibodies conjugated with short DNA oligonucleotides visualized multiple targets by sequential exchange of fluorophore-labeled complementary oligonucleotides present in the imaging buffer. This approach avoids potential effects on structural integrity when using multiple rounds of immunolabeling and eliminates chromatic aberration, because all targets are imaged using a single excitation laser wavelength. This method proved robust for multi-target imaging in semi-thin tissue sections with a lateral resolution better than 25 nm, paving the way toward structural cell biology with single-molecule SRM.
Collapse
Affiliation(s)
- Kaarjel K Narayanasamy
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Aleksandar Stojic
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Yunqing Li
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Steffen Sass
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Marina R Hesse
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Nina S Deussner-Helfmann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Marina S Dietz
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Maja Klevanski
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Mike Heilemann
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
16
|
Bobić-Rasonja M, Pogledić I, Mitter C, Štajduhar A, Milković-Periša M, Trnski S, Bettelheim D, Hainfellner JA, Judaš M, Prayer D, Jovanov-Milošević N. Developmental Differences Between the Limbic and Neocortical Telencephalic Wall: An Intrasubject Slice-Matched 3 T MRI-Histological Correlative Study in Humans. Cereb Cortex 2021; 31:3536-3550. [PMID: 33704445 DOI: 10.1093/cercor/bhab030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
The purpose of the study was to investigate the interrelation of the signal intensities and thicknesses of the transient developmental zones in the cingulate and neocortical telencephalic wall, using T2-weighted 3 T-magnetic resonance imaging (MRI) and histological scans from the same brain hemisphere. The study encompassed 24 postmortem fetal brains (15-35 postconceptional weeks, PCW). The measurements were performed using Fiji and NDP.view2. We found that T2w MR signal-intensity curves show a specific regional and developmental stage profile already at 15 PCW. The MRI-histological correlation reveals that the subventricular-intermediate zone (SVZ-IZ) contributes the most to the regional differences in the MRI-profile and zone thicknesses, growing by a factor of 2.01 in the cingulate, and 1.78 in the neocortical wall. The interrelations of zone or wall thicknesses, obtained by both methods, disclose a different rate and extent of shrinkage per region (highest in neocortical subplate and SVZ-IZ) and stage (highest in the early second half of fetal development), distorting the zones' proportion in histological sections. This intrasubject, slice-matched, 3 T correlative MRI-histological study provides important information about regional development of the cortical wall, critical for the design of MRI criteria for prenatal brain monitoring and early detection of cortical or other brain pathologies in human fetuses.
Collapse
Affiliation(s)
- Mihaela Bobić-Rasonja
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, 10000 Zagreb, Croatia.,Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivana Pogledić
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Christian Mitter
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Andrija Štajduhar
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, 10000 Zagreb, Croatia.,Andrija Štampar School of Public Health, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marija Milković-Periša
- University Hospital Centre Zagreb, Department of Pathology and Cytology, 10000 Zagreb, Croatia
| | - Sara Trnski
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, 10000 Zagreb, Croatia
| | - Dieter Bettelheim
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes A Hainfellner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Miloš Judaš
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, 10000 Zagreb, Croatia
| | - Daniela Prayer
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Nataša Jovanov-Milošević
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, 10000 Zagreb, Croatia.,Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
17
|
Excitatory/Inhibitory Synaptic Ratios in Polymicrogyria and Down Syndrome Help Explain Epileptogenesis in Malformations. Pediatr Neurol 2021; 116:41-54. [PMID: 33450624 DOI: 10.1016/j.pediatrneurol.2020.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND The ratio between excitatory (glutamatergic) and inhibitory (GABAergic) inputs into maturing individual cortical neurons influences their epileptic potential. Structural factors during development that alter synaptic inputs can be demonstrated neuropathologically. Increased mitochondrial activity identifies neurons with excessive discharge rates. METHODS This study focuses on the neuropathological examinaion of surgical resections for epilepsy and at autopsy, in fetuses, infants, and children, using immunocytochemical markers, and electron microscopy in selected cases. Polymicrogyria and Down syndrome are highlighted. RESULTS Factors influencing afferent synaptic ratios include the following: (1) synaptic short-circuitry in fused molecular zones of adjacent gyri (polymicrogyria); (2) impaired development of dendritic spines decreasing excitation (Down syndrome); (3) extracellular keratan sulfate proteoglycan binding to somatic membranes but not dendritic spines may be focally diminished (cerebral atrophy, schizencephaly, lissencephaly, polymicrogyria) or augmented, ensheathing individual axons (holoprosencephaly), or acting as a barrier to axonal passage in the U-fiber layer. If keratan is diminished, glutamate receptors on the neuronal soma enable ectopic axosomatic excitatory synapses to form; (4) dysplastic, megalocytic neurons and balloon cells in mammalian target of rapamycin disorders; (5) satellitosis of glial cells displacing axosomatic synapses; (6) peri-neuronal inflammation (tuberous sclerosis) and heat-shock proteins. CONCLUSIONS Synaptic ratio of excitatory/inhibitory afferents is a major fundamental basis of epileptogenesis at the neuronal level. Neuropathology can demonstrate subcellular changes that help explain either epilepsy or lack of seizures in immature brains. Synaptic ratios in malformations influence postnatal epileptogenesis. Single neurons can be hypermetabolic and potentially epileptogenic.
Collapse
|
18
|
Loss of the fragile X syndrome protein FMRP results in misregulation of nonsense-mediated mRNA decay. Nat Cell Biol 2021; 23:40-48. [PMID: 33420492 PMCID: PMC8273690 DOI: 10.1038/s41556-020-00618-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/29/2020] [Indexed: 01/28/2023]
Abstract
Loss of the fragile X protein FMRP is a leading cause of intellectual disability and autism1,2, but the underlying mechanism remains poorly understood. We report that FMRP deficiency results in hyperactivated nonsense-mediated mRNA decay (NMD)3,4 in human SH-SY5Y neuroblastoma cells and fragile X syndrome (FXS) fibroblast-derived induced pluripotent stem cells (iPSCs). We examined the underlying mechanism and found that the key NMD factor UPF1 binds directly to FMRP, promoting FMRP binding to NMD targets. Our data indicate that FMRP acts as an NMD repressor. In the absence of FMRP, NMD targets are relieved from FMRP-mediated translational repression so that their half-lives are decreased and, for those NMD targets encoding NMD factors, increased translation produces abnormally high factor levels despite their hyperactivated NMD. Transcriptome-wide alterations caused by NMD hyperactivation have a role in the FXS phenotype. Consistent with this, small-molecule-mediated inhibition of hyperactivated NMD, which typifies iPSCs derived from patients with FXS, restores a number of neurodifferentiation markers, including those not deriving from NMD targets. Our mechanistic studies reveal that many molecular abnormalities in FMRP-deficient cells are attributable-either directly or indirectly-to misregulated NMD.
Collapse
|
19
|
Tanneti NS, Federspiel JD, Cristea IM, Enquist LW. The axonal sorting activity of pseudorabies virus Us9 protein depends on the state of neuronal maturation. PLoS Pathog 2020; 16:e1008861. [PMID: 33370419 PMCID: PMC7794026 DOI: 10.1371/journal.ppat.1008861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/08/2021] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
Alpha-herpesviruses establish a life-long infection in the nervous system of the affected host; while this infection is restricted to peripheral neurons in a healthy host, the reactivated virus can spread within the neuronal circuitry, such as to the brain, in compromised individuals and lead to adverse health outcomes. Pseudorabies virus (PRV), an alpha-herpesvirus, requires the viral protein Us9 to sort virus particles into axons and facilitate neuronal spread. Us9 sorts virus particles by mediating the interaction of virus particles with neuronal transport machinery. Here, we report that Us9-mediated regulation of axonal sorting also depends on the state of neuronal maturation. Specifically, the development of dendrites and axons is accompanied with proteomic changes that influence neuronal processes. Immature superior cervical ganglionic neurons (SCGs) have rudimentary neurites that lack markers of mature axons. Immature SCGs can be infected by PRV, but they show markedly reduced Us9-dependent regulation of sorting, and increased Us9-independent transport of particles into neurites. Mature SCGs have relatively higher abundances of proteins characteristic of vesicle-transport machinery. We also identify Us9-associated neuronal proteins that can contribute to axonal sorting and subsequent anterograde spread of virus particles in axons. We show that SMPD4/nsMase3, a sphingomyelinase abundant in lipid-rafts, associates with Us9 and is a negative regulator of PRV sorting into axons and neuronal spread, a potential antiviral function.
Collapse
Affiliation(s)
- Nikhila S. Tanneti
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Joel D. Federspiel
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Lynn W. Enquist
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
20
|
Wei H, Zhou W, Hu G, Shi C. Induction of mesenchymal stem cell‑like transformation in rat primary glial cells using hypoxia, mild hypothermia and growth factors. Mol Med Rep 2020; 23:121. [PMID: 33300053 PMCID: PMC7751450 DOI: 10.3892/mmr.2020.11760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023] Open
Abstract
The transformation of rat primary glial cells into mesenchymal stem cells (MSCs) is intriguing as more seed cells can be harvested. The present study aimed to evaluate the effects of growth factors, hypoxia and mild hypothermia on the transformation of primary glial cells into MSCs. Rat primary glial cells were induced to differentiate by treatment with hypoxia, mild hypothermia and basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF). Immunohistochemistry and western blotting were then used to determine the expression levels of glial fibrillary acidic protein (GFAP), nestin, musashi-1, neuron specific enolase (NSE) and neuronal nuclei (NeuN), in each treatment group. bFGF and EGF increased the proportion of CD44+ and CD105+ cells, while anaerobic mild hypothermia increased the proportion of CD90+ cells. The combination of bFGF and EGF, and anaerobic mild hypothermia increased the proportion of CD29+ cells and significantly decreased the proportions of GFAP+ cells and NSE+ cells. Treatment of primary glial cells with bFGF and EGF increased the expression levels of nestin, Musashi-1, NSE and NeuN. Anaerobic mild hypothermia increased the expression levels of Musashi-1 and decreased the expression levels of NSE and NeuN in glial cells. The results of the present study demonstrated that bFGF, EGF and anaerobic mild hypothermia treatments may promote the transformation of glial cells into MSC-like cells, and that the combination of these two treatments may have the optimal effect.
Collapse
Affiliation(s)
- Huiping Wei
- Department of Health Care for Cadres, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenyun Zhou
- Department of Prevention and Health Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guozhu Hu
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chunhua Shi
- Department of Rheumatology and Immunology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
21
|
Sarnat Grading Scale for Neonatal Encephalopathy after 45 Years: An Update Proposal. Pediatr Neurol 2020; 113:75-79. [PMID: 33069006 DOI: 10.1016/j.pediatrneurol.2020.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 11/22/2022]
|
22
|
Radix Scutellariae Ameliorates Stress-Induced Depressive-Like Behaviors via Protecting Neurons through the TGF β3-Smad2/3-Nedd9 Signaling Pathway. Neural Plast 2020; 2020:8886715. [PMID: 33273910 PMCID: PMC7683137 DOI: 10.1155/2020/8886715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/29/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022] Open
Abstract
Chronic stress can impair hippocampal neurogenesis, increase neuronal apoptosis, and cause depressive-like behaviors. Our previous studies found that Radix Scutellariae (RS) can rescue the stress-induced neuronal injury, but the mechanism is not clear. Here, we continued to investigate the underlying antidepressant mechanisms of the RS extract. A 7-week chronic unpredictable mild stress (CUMS) procedure was used to establish a murine depression model. 0.75 g/kg or 1.5 g/kg RS was administered daily to the mice during the last 4 weeks. Depressive-like behaviors were evaluated by the sucrose preference test (SPT), forced swimming test (FST), open field test (OFT), and tail suspension test (TST). The neuroprotective effect of RS was evaluated with the expression of hippocampal neuron-related markers and apoptosis-associated proteins by Nissl staining, immunohistochemistry, and western blot. Transforming growth factor-β3 (TGFβ3) pathway-related proteins were detected by western blot. Results showed that RS could ameliorate depressive-like behaviors, increase the expression of the antiapoptotic protein B-cell lymphoma 2 (BCL-2), reduce the expression of the proapoptotic protein BCL-2-associated X (BAX), and increase the number of doublecortin- (DCX-), microtubule-associated protein 2- (MAP2-), and neuronal nucleus- (NeuN-) positive cells in the hippocampus. Moreover, RS could reverse the CUMS-induced decrease of TGFβ3 protein, promote the phosphorylation of SMAD2/3, and increase the expression of downstream NEDD9 protein. These results suggest that RS could exert antidepressant effects via protecting neurons. And the molecular mechanism might be related to the regulation of the TGFβ3-SMAD2/3-NEDD9 pathway.
Collapse
|
23
|
Sarnat HB. Proteoglycan (Keratan Sulfate) Barrier in Developing Human Forebrain Isolates Cortical Epileptic Networks From Deep Heterotopia, Insulates Axonal Fascicles, and Explains Why Axosomatic Synapses Are Inhibitory. J Neuropathol Exp Neurol 2020; 78:1147-1159. [PMID: 31633782 DOI: 10.1093/jnen/nlz096] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Axons from deep heterotopia do not extend through U-fibers, except transmantle dysplasias. Keratan sulfate (KS) in fetal spinal cord/brainstem median septum selectively repels glutamatergic axons while enabling GABAergic commissural axons. Immunocytochemical demonstration of KS in neocortical resections and forebrain at autopsy was studied in 12 fetuses and neonates 9-41 weeks gestational age (GA), 9 infants, children, and adolescents and 5 patients with focal cortical dysplasias (FCD1a). From 9 to 15 weeks GA, no KS is seen in the cortical plate; 19-week GA reactivity is detected in the molecular zone. By 28 weeks GA, patchy granulofilamentous reactivity appears in extracellular matrix and adheres to neuronal somata with increasing intensity in deep cortex and U-fibers at term. Perifascicular KS surrounds axonal bundles of both limbs of the internal capsule and within basal ganglia from 9 weeks GA. Thalamus and globus pallidus exhibit intense astrocytic reactivity from 9 weeks GA. In FCD1a, U-fiber reactivity is normal, discontinuous or radial. Ultrastructural correlates were not demonstrated; KS is not electron-dense. Proteoglycan barrier of the U-fiber layer impedes participation of deep heterotopia in cortical epileptic networks. Perifascicular KS prevents aberrant axonal exit from or entry into long and short tracts. KS adhesion to neuronal somatic membranes may explain inhibitory axosomatic synapses.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics, Pathology (Neuropathology), and Clinical Neurosciences, University of Calgary, Cumming School of Medicine; and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, Alberta, Canada
| |
Collapse
|
24
|
Gilani A, Hove JLV, Thomas JA, Kleinschmidt-DeMasters BK. Distinguishing Encephaloclastic Lesions Resulting From Primary or Secondary Pyruvate Dehydrogenase Deficiency From Other Neonatal or Infantile Cavitary Brain Lesions. Pediatr Dev Pathol 2020; 23:189-196. [PMID: 31542992 DOI: 10.1177/1093526619876448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The central nervous system (CNS) is a highly complex and energy-dependent organ that is subject to a wide variety of metabolic, hypoxic-ischemic, and infectious insults that result in cystic changes. Diagnosis of metabolic defects causing extensive cystic changes is particularly challenging for the pediatric pathologist, due to the rarity of these conditions. Pyruvate dehydrogenase (PDH) deficiency is one of the most common etiologies of congenital lactic acidosis, caused by mutations in subunits of the large mitochondrial matrix complex, and characterized by periventricular cysts, although few detailed reports focusing on neuropathologic findings exist. In addition, rare defects in other mitochondrial enzymes such as short-chain enoyl-CoA hydratase (SCEH, encoded by ECHS1 gene) can cause secondary PDH deficiency and present with neonatal lactic acidosis, but neuropathological findings have never been reported. Nonmetabolic conditions can also produce CNS cystic lesions, primarily in newborns. The pathologist must therefore distinguish between these etiologically disparate conditions which can produce CNS cavitary lesions. Here, we compare and contrast the gross and microscopic findings of cysts associated with cases of PDH and SCEH deficiencies with other neonatal cystic brain diseases including periventricular leukomalacia, neonatal Alexander disease, Canavan disease, and a case of cysts associated with a vascular abnormality. Our studies show that PDH and SCEH deficiencies are not grossly or histologically distinguishable from each other and both are associated with smooth-walled cysts largely limited to the telencephalic germinal matrix. Both show an absence of prominent hemosiderin deposits, Rosenthal fibers, vacuolization of the white matter, and gliosis or axonal damage in the surrounding parenchyma. These features can help distinguish PDH/SCEH deficiency from other pediatric/neonatal cystic CNS disorders, especially those produced by hypoxic ischemic conditions. Cysts, usually bilateral, confined to the telencephalic germinal matrix should elicit metabolic and genetic testing to appropriately diagnose PDH and SCEH and distinguish them from each other.
Collapse
Affiliation(s)
- Ahmed Gilani
- Department of Pathology, Children's Hospital Colorado, University of Colorado, Aurora, Colorado
| | - Johan Lk Van Hove
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado
| | - Janet A Thomas
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado
| | | |
Collapse
|
25
|
Kim B, De La Monte S, Hovanesian V, Patra A, Chen X, Chen RH, Miller MC, Pinar MH, Lim YP, Stopa EG, Stonestreet BS. Ontogeny of inter-alpha inhibitor protein (IAIP) expression in human brain. J Neurosci Res 2019; 98:869-887. [PMID: 31797408 DOI: 10.1002/jnr.24565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 12/19/2022]
Abstract
Inter-alpha inhibitor proteins (IAIPs) are naturally occurring immunomodulatory molecules found in most tissues. We have reported ontogenic changes in the expression of IAIPs in brain during development in sheep and abundant expression of IAIPs in fetal and neonatal rodent brain in a variety of cellular types and brain regions. Although a few studies identified bikunin, light chain of IAIPs, in adult human brain, the presence of the complete endogenous IAIP protein complex has not been reported in human brain. In this study, we examined the immunohistochemical expression of endogenous IAIPs in human cerebral cortex from early in development through the neonatal period and in adults using well-preserved postmortem brains. We examined total, nuclear, and cytoplasmic staining of endogenous IAIPs and their expression in neurofilament light polypeptide-positive neurons and glial fibrillary acidic protein (GFAP)-positive astrocytes. IAIPs were ubiquitously detected for the first time in cerebral cortical cells at 24-26, 27-28, 29-36, and 37-40 weeks of gestation and in adults. Quantitative analyses revealed that IAIPs were predominately localized in the nucleus in all age groups, but cytoplasmic IAIP expression was more abundant in adult than in the younger ages. Immunoreactivity of IAIPs was expressed in neurons and astrocytes in all age groups. In addition, IAIP co-localization with GFAP-positive astrocytes was more abundant in adults than in the developing brain. We conclude that IAIPs exhibit ubiquitous expression, and co-localize with neurons and astrocytes in the developing and adult human brain suggesting a potential role for IAIPs in development and endogenous neuroprotection.
Collapse
Affiliation(s)
- Boram Kim
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Suzanne De La Monte
- Department of Neurology and Neurosurgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Aparna Patra
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Xiaodi Chen
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Ray H Chen
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Miles C Miller
- Department of Pathology and Neurosurgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Mehmet Halit Pinar
- Department of Pathology & Laboratory Medicine, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Yow-Pin Lim
- Department of Pathology & Laboratory Medicine, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA.,ProThera Biologics, Inc., Providence, RI, USA
| | - Edward G Stopa
- Department of Pathology and Neurosurgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| |
Collapse
|
26
|
Bárez-López S, Grijota-Martínez C, Ausó E, Fernández-de Frutos M, Montero-Pedrazuela A, Guadaño-Ferraz A. Adult Mice Lacking Mct8 and Dio2 Proteins Present Alterations in Peripheral Thyroid Hormone Levels and Severe Brain and Motor Skill Impairments. Thyroid 2019; 29:1669-1682. [PMID: 31359845 DOI: 10.1089/thy.2019.0068] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Mutations in the thyroid hormone (TH) transporter monocarboxylate transporter 8 (MCT8) lead to peripheral hyperthyroidism and profound psychomotor alterations in humans. Mice lacking Mct8 present peripheral hyperthyroidism but no gross neurological abnormalities due to brain compensatory mechanisms involving the enzyme deiodinase type 2 (Dio2). Methods: Here we have analyzed the endocrine and neurologic phenotype of mice lacking both Mct8 and Dio2 at three and six months of age. Thyroxine (T4) and 3,5,3' triiodothyronine (T3) levels/content were measured by specific radioimmunoassays; motor skill performance was evaluated by the footprint, rotarod, four limb hanging wire, and balance beam tests; and brain histological analysis was performed by immunostaining for neurofilament and parvalbumin. Results: We have found that this mouse model presents peripheral hyperthyroidism and brain hypothyroidism. Interestingly, the severity of the brain hypothyroidism seems permanent and varies across regions, with the striatum being a particularly affected area. We have also found brain alterations at the histological level compatible with TH deficiency and impaired motor skills. Conclusions: These findings indicate the potential of Mct8/Dio2-deficient mice to represent a model for human MCT8 deficiency, to understand the mechanisms underlying its pathophysiology, and ultimately design therapeutic interventions for human patients.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Unit 708, Center for Biomedical Research On Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Grijota-Martínez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Unit 708, Center for Biomedical Research On Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva Ausó
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, Alicante, Spain
| | - Mario Fernández-de Frutos
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Montero-Pedrazuela
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Unit 708, Center for Biomedical Research On Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
27
|
Sarnat HB, Flores-Sarnat L, Boltshauser E. Area Postrema: Fetal Maturation, Tumors, Vomiting Center, Growth, Role in Neuromyelitis Optica. Pediatr Neurol 2019; 94:21-31. [PMID: 30797593 DOI: 10.1016/j.pediatrneurol.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The area postrema in the caudal fourth ventricular floor is highly vascular without blood-brain or blood-cerebrospinal fluid barrier. In addition to its function as vomiting center, several others are part of the circumventricular organs for vasomotor/angiotensin II regulation, role in neuromyelitis optica related to aquaporin-4, and somatic growth and appetite regulation. Functions are immature at birth. The purpose was to demonstrate neuronal, synaptic, glial, or ependymal maturation in the area postrema of normal fetuses. We describe three area postrema tumors. METHODS Sections of caudal fourth ventricle of 12 normal human fetal brains at autopsy aged six to 40 weeks and three infants aged three to 18 months were examined. Immunocytochemical neuronal and glial markers were applied to paraffin sections. Two infants with area postrema tumors and another with neurocutaneous melanocytosis and pernicious vomiting also studied. RESULTS Area postrema neurons exhibited cytologic maturity and synaptic circuitry by 14 weeks'. Astrocytes coexpressed vimentin, glial fibrillary acidic protein, and S-100β protein. The ependyma is thin over area postrema, with fetal ependymocytic basal processes. A glial layer separates area postrema from medullary tegmentum. Melanocytes infiltrated area postrema in the toddler with pernicious vomiting; two children had primary area postrema pilocytic astrocytomas. CONCLUSIONS Although area postrema is cytologically mature by 14 weeks, growth increases and functions mature during postnatal months. We recommend neuroimaging for patients with unexplained vomiting and that area postrema neuropathology includes synaptophysin and microtubule-associated protein-2 in patients with suspected dysfunction.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Pathology (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.
| | - Laura Flores-Sarnat
- Departments of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Eugen Boltshauser
- Department of Paediatric Neurology, Children's University Hospital, Zürich, Switzerland
| |
Collapse
|
28
|
Taylor MA, Kan HL, Gollapudi BB, Marty MS. An in vitro developmental neurotoxicity screening assay for retinoic acid-induced neuronal differentiation using the human NT2/D1 cell line. Neurotoxicology 2019; 73:258-264. [PMID: 30980846 DOI: 10.1016/j.neuro.2019.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/04/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
Abstract
Traditional approaches (e.g., neurobehavior, neuropathology) can detect alterations in apical endpoints indicative of developmental neurotoxicity (DNT). However, there is an increasing desire to understand mode-of-action (MOA) for DNT effects; thus, this short communication describes initial work on a neuronal differentiation assay. Basically, our laboratory used the human NT2/D1 cell line to develop an assay to evaluate toxicants for effects on all-trans retinoic acid (RA)-induced neuronal differentiation. Based on literature reports, we selected a neuronal protein, neuronal class III β-tubulin (β3-tubulin), as a marker of differentiation. For this assay, cultured RA-treated NT2 cells were trypsinized to individual cells, methanol fixed, and labeled with a β3-tubulin specific monoclonal antibody (TUJ1). Characterization studies using 100,000 cells/sample showed that NT2 cells had appreciable expression of β3-tubulin starting around day 7 of the differentiation process with a peak expression noted around day 12. Methylmercury, 22(R)-hydroxycholesterol, N-(4-hydroxyphenol)retinamide (4HPR), and 9-cis retinoic acid were selected as initial test compounds. Of these, only 9-cis RA, which is known to affect the RA pathway, was positive for specific impacts on differentiation. These results demonstrate the feasibility of using a flow cytometry method targeting specific cellular biomarkers for evaluating effects on neuronal differentiation. Additional assays are needed to detect compounds targeting other (non-RA) neuronal differentiation pathways. Ultimately, a battery of in vitro assays would be needed to evaluate the potential MOAs involved in altered neuronal differentiation.
Collapse
Affiliation(s)
| | - H Lynn Kan
- The Dow Chemical Company, Midland, MI, USA
| | | | | |
Collapse
|
29
|
Mühlebner A, Bongaarts A, Sarnat HB, Scholl T, Aronica E. New insights into a spectrum of developmental malformations related to mTOR dysregulations: challenges and perspectives. J Anat 2019; 235:521-542. [PMID: 30901081 DOI: 10.1111/joa.12956] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
In recent years the role of the mammalian target of rapamycin (mTOR) pathway has emerged as crucial for normal cortical development. Therefore, it is not surprising that aberrant activation of mTOR is associated with developmental malformations and epileptogenesis. A broad spectrum of malformations of cortical development, such as focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC), have been linked to either germline or somatic mutations in mTOR pathway-related genes, commonly summarised under the umbrella term 'mTORopathies'. However, there are still a number of unanswered questions regarding the involvement of mTOR in the pathophysiology of these abnormalities. Therefore, a monogenetic disease, such as TSC, can be more easily applied as a model to study the mechanisms of epileptogenesis and identify potential new targets of therapy. Developmental neuropathology and genetics demonstrate that FCD IIb and hemimegalencephaly are the same diseases. Constitutive activation of mTOR signalling represents a shared pathogenic mechanism in a group of developmental malformations that have histopathological and clinical features in common, such as epilepsy, autism and other comorbidities. We seek to understand the effect of mTOR dysregulation in a developing cortex with the propensity to generate seizures as well as the aftermath of the surrounding environment, including the white matter.
Collapse
Affiliation(s)
- A Mühlebner
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Bongaarts
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H B Sarnat
- Departments of Paediatrics, Pathology (Neuropathology) and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, AB, Canada
| | - T Scholl
- Department of Paediatric and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - E Aronica
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Amsterdam, The Netherlands
| |
Collapse
|
30
|
Abstract
This chapter focuses on the development of the human olfactory system. In this system, function does not require full neuroanatomical maturity. Thus, discrimination of odorous molecules, including a number within the mother's diet, occurs in amniotic fluid after 28-30 weeks of gestation, at which time the olfactory bulbs are identifiable by MRI. Hypoplasia/aplasia of the bulbs is documented in the third trimester and postnatally. Interestingly, olfactory axons project from the nasal epithelium to the telencephalon before formation of the olfactory bulbs and lack a peripheral ganglion, but the synaptic glomeruli of the future olfactory bulb serves this function. Histologic lamination of the olfactory bulb is present by 14 weeks, but maturation remains incomplete at term for neuronal differentiation, synaptogenesis, myelination, and persistence of the normal transitory fetal ventricular recess. Myelination occurs postnatally. Although olfaction is the only sensory system without direct thalamic projections, the olfactory bulb and anterior olfactory nucleus are, in effect, thalamic surrogates. For example, many dendro-dendritic synapses occur within the bulb between GABAergic granular neurons and periglomerular neurons. Moreover, bulbar synaptic glomeruli are analogous to peripheral ganglia of other sensory cranial nerves. The olfactory tract contains much gray as well as white matter. The olfactory epithelium and bulb both incorporate progenitor cells at all ages. Diverse malformations of the olfactory bulb can be detected by clinical examination, imaging, and neuropathology; indeed, olfactory reflexes of the neonate can be reliably tested. We recommend that such testing be routine in the neonatal neurologic examination, especially in children with brain malformations, endocrinopathies, chromosomopathies, genetic/metabolic disorders, and perinatal hypoxic/ischemic encephalopathy.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Department of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, AB, Canada; Department of Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, AB, Canada; Department of Pathology and Laboratory Medicine (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, AB, Canada.
| | - Laura Flores-Sarnat
- Department of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, AB, Canada; Department of Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, AB, Canada
| |
Collapse
|
31
|
Shah FA, Zeb A, Ali T, Muhammad T, Faheem M, Alam SI, Saeed K, Koh PO, Lee KW, Kim MO. Identification of Proteins Differentially Expressed in the Striatum by Melatonin in a Middle Cerebral Artery Occlusion Rat Model-a Proteomic and in silico Approach. Front Neurosci 2018; 12:888. [PMID: 30618542 PMCID: PMC6295458 DOI: 10.3389/fnins.2018.00888] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke is characterized by permanent or transient obstruction of blood flow, which initiates a cascading pathological process, starting from acute ATP loss to subsequent membrane depolarization, glutamate excitotoxicity, and calcium overload. Melatonin is a potent antioxidant that exerts protective effects in different experimental stroke models. In this study, melatonin effects were demonstrated by a proteomic and in silico approach. The proteomic study identified differentially expressed proteins by 2D gel electrophoresis in the striatum 24 h after middle cerebral artery occlusion. Proteomic analysis revealed several proteins with aberrant expression and was validated by western blot and immunofluorescence analysis. Homology modeling was performed to build 3D structures for γ-enolase, thioredoxin (TRX), and heat shock 60 (HSP60) by the template crystal structures using a protein data bank as a sequence database. The structure refinement of each model was achieved by energy minimization via molecular dynamic simulation, and the generated models were further assessed for stability by Procheck and ProSA. The models were processed for docking analysis using AutoDock Vina, and post-docking analysis was determined by discovery studio. The proteomic study showed decreased expression of γ-enolase, TRX, and protein phosphatase 2A subunit B and increased expression of collapsin response mediator protein 2 and HSP60 in the striatum after ischemic injury. Treatment with melatonin modulated the expression profiles of these proteins. This study demonstrated the neuroprotective role of melatonin in the ischemic striatum using a proteomic and in silico approach. Collectively, melatonin may act in a multimechanistic way by modulating the expression of several proteins in the ischemic striatum.
Collapse
Affiliation(s)
- Fawad Ali Shah
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea.,Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad, Rawalpindi, Pakistan
| | - Amir Zeb
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Tahir Ali
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea.,Department of Pharmacy, Faculty of Life Science, Sarhad University of Science and Information Technology, Peshawar, Pakistan
| | - Tahir Muhammad
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Muhammad Faheem
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad, Rawalpindi, Pakistan
| | - Sayed Ibrar Alam
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Kamran Saeed
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Keun Woo Lee
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
32
|
Žunić Išasegi I, Radoš M, Krsnik Ž, Radoš M, Benjak V, Kostović I. Interactive histogenesis of axonal strata and proliferative zones in the human fetal cerebral wall. Brain Struct Funct 2018; 223:3919-3943. [PMID: 30094607 PMCID: PMC6267252 DOI: 10.1007/s00429-018-1721-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022]
Abstract
Development of the cerebral wall is characterized by partially overlapping histogenetic events. However, little is known with regards to when, where, and how growing axonal pathways interact with progenitor cell lineages in the proliferative zones of the human fetal cerebrum. We analyzed the developmental continuity and spatial distribution of the axonal sagittal strata (SS) and their relationship with proliferative zones in a series of human brains (8-40 post-conceptional weeks; PCW) by comparing histological, histochemical, and immunocytochemical data with magnetic resonance imaging (MRI). Between 8.5 and 11 PCW, thalamocortical fibers from the intermediate zone (IZ) were initially dispersed throughout the subventricular zone (SVZ), while sizeable axonal "invasion" occurred between 12.5 and 15 PCW followed by callosal fibers which "delaminated" the ventricular zone-inner SVZ from the outer SVZ (OSVZ). During midgestation, the SS extensively invaded the OSVZ, separating cell bands, and a new multilaminar axonal-cellular compartment (MACC) was formed. Preterm period reveals increased complexity of the MACC in terms of glial architecture and the thinning of proliferative bands. The addition of associative fibers and the formation of the centrum semiovale separated the SS from the subplate. In vivo MRI of the occipital SS indicates a "triplet" structure of alternating hypointense and hyperintense bands. Our results highlighted the developmental continuity of sagittally oriented "corridors" of projection, commissural and associative fibers, and histogenetic interaction with progenitors, neurons, and glia. Histogenetical changes in the MACC, and consequently, delineation of the SS on MRI, may serve as a relevant indicator of white matter microstructural integrity in the developing brain.
Collapse
Affiliation(s)
- Iris Žunić Išasegi
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Milan Radoš
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Marko Radoš
- Department of Radiology, Clinical Hospital Center Zagreb, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Vesna Benjak
- Department of Pediatrics, Clinical Hospital Center Zagreb, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia.
| |
Collapse
|
33
|
Trattnig C, Üçal M, Tam-Amersdorfer C, Bucko A, Zefferer U, Grünbacher G, Absenger-Novak M, Öhlinger KA, Kraitsy K, Hamberger D, Schaefer U, Patz S. MicroRNA-451a overexpression induces accelerated neuronal differentiation of Ntera2/D1 cells and ablation affects neurogenesis in microRNA-451a-/- mice. PLoS One 2018; 13:e0207575. [PMID: 30462722 PMCID: PMC6248975 DOI: 10.1371/journal.pone.0207575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/02/2018] [Indexed: 12/22/2022] Open
Abstract
MiR-451a is best known for its role in erythropoiesis and for its tumour suppressor features. Here we show a role for miR-451a in neuronal differentiation through analysis of endogenous and ectopically expressed or silenced miR-451a in Ntera2/D1 cells during neuronal differentiation. Furthermore, we compared neuronal differentiation in the dentate gyrus of hippocampus of miR-451a-/- and wild type mice. MiR-451a overexpression in lentiviral transduced Ntera2/D1 cells was associated with a significant shifting of mRNA expression of the developmental markers Nestin, βIII Tubulin, NF200, DCX and MAP2 to earlier developmental time points, compared to control vector transduced cells. In line with this, accelerated neuronal network formation in AB.G.miR-451a transduced cells, as well as an increase in neurite outgrowth both in number and length was observed. MiR-451a targets genes MIF, AKT1, CAB39, YWHAZ, RAB14, TSC1, OSR1, POU3F2, TNS4, PSMB8, CXCL16, CDKN2D and IL6R were, moreover, either constantly downregulated or exhibited shifted expression profiles in AB.G.miR-451a transduced cells. Lentiviral knockdown of endogenous miR-451a expression in Ntera2/D1 cells resulted in decelerated differentiation. Endogenous miR-451a expression was upregulated during development in the hippocampus of wildtype mice. In situ hybridization revealed intensively stained single cells in the subgranular zone and the hilus of the dentate gyrus of wild type mice, while genetic ablation of miR-451a was observed to promote an imbalance between proliferation and neuronal differentiation in neurogenic brain regions, suggested by Ki67 and DCX staining. Taken together, these results provide strong support for a role of miR-451a in neuronal maturation processes in vitro and in vivo.
Collapse
Affiliation(s)
- Christa Trattnig
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | - Muammer Üçal
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | | | - Angela Bucko
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | - Ulrike Zefferer
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | - Gerda Grünbacher
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | | | | | - Klaus Kraitsy
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | - Daniel Hamberger
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| | - Ute Schaefer
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
- * E-mail:
| | - Silke Patz
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University, Graz, Austria
| |
Collapse
|
34
|
Özbek M, Bozkurt MF, Beyaz F, Ergün E, Ergün L. Expression profile of some neuronal and glial cell markers in the ovine ileal enteric nervous system during prenatal development. Acta Histochem 2018; 120:768-779. [PMID: 30217408 DOI: 10.1016/j.acthis.2018.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/07/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
The enteric nervous system (ENS) is a network of neurons and glia found in the gut wall and governs this gastrointestinal function independently from the central nervous system (CNS). ENS comprises the myenteric plexus (MP) and the submucous plexus (SP). In this study, we examined the expression profile of neurofilament heavy chain (NF-H), neuron-specific enolase (NSE), calcyclin (S100A6), vimentin and glial fibril acidic protein (GFAP) in ovine ileal enteric neurons and enteric glia cells (EGCs) during prenatal development using an immunohistochemical method. The material of the study consisted of 15 different fetal ileum tissues obtained between days 60 and 150 of pregnancy. NF-H was observed in the majority of ganglion cells in SP and MP throughout the fetal period. It was determined that there was no NF-H reaction in some ganglion cells in Peyer's patches of internal submucosal plexus (ISPF). In the early stage of pregnancy (60-90 days), there was no expression of NSE and S1006 in ileum. After this period, NSE and S1006 were expressed in the ganglion cells of the plexus, indicating an increase in the amount of expression towards the end of pregnancy. In the early period, vimentin expression was only detected in intramuscular interstitial cells (ICs) (60-90 days), but later (90-150 days) it was also seen in the cells around the ganglion cells in the plexus. On days 60-90 of gestation, GFAP expression only occurred in MP, but in later stages, staining was also detected in SP. In the plexus, an immunoreactivity was present in EGCs forming a network around the ganglion cell. During the last period of gestation (120-150 days), the number of GFAP-positive plexus increased, with the majority of these stained cells being observed in MP. Interestingly, weak staining or reaction did not occur in ISPF, unlike other plexuses. In conclusion, this is the first study that demonstrated the expression of NF-H, vimentin, S100A6, NSE and glial fibril acidic protein (GFAP) in ovine ileal ENS in the prenatal period. In the last period of gestation (120-150 days), the expression profile of ENS was similar to that of adult animals. The expression of the used markers increased toward the end of pregnancy. Our results suggest that neurons and EGCs show heterogeneity, and GFAP and NF-H cannot be used as panenteric glial or panneuronal markers, respectively. We also demonstrated, for the first time, the prenatal expression of S100A6 in enteric neurons and the possibility of using this protein for the identification of enteric neurons.
Collapse
Affiliation(s)
- Mehmet Özbek
- Deparment of Histology and Embryology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, İstiklal Yerleşkesi, 15030, Burdur, Turkey.
| | - Mehmet Fatih Bozkurt
- Department of Pathology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Feyzullah Beyaz
- Deparment of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Emel Ergün
- Deparment of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Levent Ergün
- Deparment of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
35
|
Grigsby KB, Kelty TJ, Booth FW. Medial habenula maturational deficits associate with low motivation for voluntary physical activity. Brain Res 2018; 1698:187-194. [PMID: 30118717 DOI: 10.1016/j.brainres.2018.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/11/2018] [Accepted: 08/13/2018] [Indexed: 11/25/2022]
Abstract
The habenula is a small, diencephalic structure comprised of distinct subnuclei which receives inputs from the limbic forebrain and sends projections to various regions in the midbrain, making this region well positioned to influence reward and motivation. Genetic ablation of the dorsal medial habenula is known to decrease voluntary wheel-running in mice. However, the extent to which the medial habenula (MHb) mediates wheel-running motivation in the context of high or low motivation for voluntary physical activity remains to be determined. In so, we utilized 5-week-old female rats selectively bred to voluntarily run high (HVR) or low (LVR) distances in order to determine if inherent differences in medial habenula maturation accompany inherent differences in wheel-running motivation. We report a significantly higher expression of genes associated with MHb development (Brn3a, Nurr1, Tac1, and Kcnip) in HVR versus LVR rats. Furthermore, there was a positive correlation between Brn3a and Nurr1 expression and run distance in HVR, but not LVR rats. Similarly, NeuN and Synapsin 1, markers of neuronal maturation, were higher in HVR compared to LVR rats. Lastly, dendritic density was determined to be higher in the MHb of HVR versus LVR rats, while LVR rats showed a higher percentage of thin spines, suggesting a higher prevalence of immature dendrites in LVR rats. Taken together, the above findings highlight the involvement of MHb in driving the motivation to be physically active. Given pandemic levels of global physical inactivity, the role of the MHb offers a novel potential to improve our global health.
Collapse
Affiliation(s)
- Kolter B Grigsby
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States.
| | - Taylor J Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States; Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
36
|
Shovlin S, Tropea D. Transcriptome level analysis in Rett syndrome using human samples from different tissues. Orphanet J Rare Dis 2018; 13:113. [PMID: 29996871 PMCID: PMC6042368 DOI: 10.1186/s13023-018-0857-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/27/2018] [Indexed: 01/06/2023] Open
Abstract
The mechanisms of neuro-genetic disorders have been mostly investigated in the brain, however, for some pathologies, transcriptomic analysis in multiple tissues represent an opportunity and a challenge to understand the consequences of the genetic mutation. This is the case for Rett Syndrome (RTT): a neurodevelopmental disorder predominantly affecting females that is characterised by a loss of purposeful movements and language accompanied by gait abnormalities and hand stereotypies. Although the genetic aetiology is largely associated to Methyl CpG binding protein 2 (MECP2) mutations, linking the pathophysiology of RTT and its clinical symptoms to direct molecular mechanisms has been difficult.One approach used to study the consequences of MECP2 dysfunction in patients, is to perform transcriptomic analysis in tissues derived from RTT patients or Induced Pluripotent Stem cells. The growing affordability and efficiency of this approach has led to a far greater understanding of the complexities of RTT syndrome but is also raised questions about previously held convictions such as the regulatory role of MECP2, the effects of different molecular mechanisms in different tissues and role of X Chromosome Inactivation in RTT.In this review we consider the results of a number of different transcriptomic analyses in different patients-derived preparations to unveil specific trends in differential gene expression across the studies. Although the analyses present limitations- such as the limited sample size- overlaps exist across these studies, and they report dysregulations in three main categories: dendritic connectivity and synapse maturation, mitochondrial dysfunction, and glial cell activity.These observations have a direct application to the disorder and give insights on the altered mechanisms in RTT, with implications on potential diagnostic criteria and treatments.
Collapse
Affiliation(s)
- Stephen Shovlin
- Neuropsychiatric Genetics Research Group, Trinity Translational Medicine Institute- TTMI, St James Hospital, D8, Dublin, Ireland
| | - Daniela Tropea
- Neuropsychiatric Genetics Research Group, Trinity Translational Medicine Institute- TTMI, St James Hospital, D8, Dublin, Ireland
- Trinity College Institute of Neuroscience, TCIN, Loyd Building, Dublin2, Dublin, Ireland
| |
Collapse
|
37
|
Sarnat HB. The 2016 Bernard Sachs Lecture: Timing in Morphogenesis and Genetic Gradients During Normal Development and in Malformations of the Nervous System. Pediatr Neurol 2018; 83:3-13. [PMID: 29778488 DOI: 10.1016/j.pediatrneurol.2017.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 08/13/2017] [Accepted: 08/28/2017] [Indexed: 10/17/2022]
Abstract
Nervous system development is quadradimensional. Both normal ontogenesis and developmental malformations are explained in the context of the fourth dimension, timing. Timing of the onset of either the genetic expression of a mutation or an epigenetic event that may be teratogenic is primordial in determining morphogenesis and the forms of malformations with their functional consequences. Multiple genotypes may cause similar phenotypes or a single genotype with different degrees of retained normal genetic expression may result in variable phenotypes. In this treatise, examples are presented of these principles, including both delayed and precocious maturation of processes such as synaptogenesis that may be out of synchrony with other simultaneous processes of neuronal maturation. In postzygotic somatic mosaicism, timing of onset determines not only the character but also the extent of a lesion; focal cortical dysplasia IIb and hemimegalencephaly are the same disease, both sharing activation of the mTOR pathway as the primary mechanism; the difference is timing of onset within the 33 mitotic cycles of the periventricular neuroepithelium. Genetic expression often follows gradients along the 3 axes of the neural tube. Defective gradients often can be identified by their morphological result without knowing the precise mutation. Upregulation in the vertical axis produces hyperplasia or duplication of either dorsal or ventral structures, whereas downregulation yields hypoplasia or fusion in the midline of bilateral structures. Disorders of segmentation or neuromere formation in the neural tube are increasingly recognized as another pathogenesis of cerebral dysgenesis. Our recent investigations show the participation of the U-fibre layer beneath FCD in epileptic networks because of neuronal dispersion with elaborate synaptic plexi and a barrier to deep heterotopia.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics, Pathology (Neuropathology), and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.
| |
Collapse
|
38
|
Patel D, Rathinam M, Jarvis C, Mahimainathan L, Henderson G, Narasimhan M. Role for Cystathionine γ Lyase (CSE) in an Ethanol (E)-Induced Lesion in Fetal Brain GSH Homeostasis. Int J Mol Sci 2018; 19:ijms19051537. [PMID: 29786653 PMCID: PMC5983808 DOI: 10.3390/ijms19051537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/19/2018] [Accepted: 05/19/2018] [Indexed: 02/06/2023] Open
Abstract
Earlier, we reported that gestational ethanol (E) can dysregulate neuron glutathione (GSH) homeostasis partially via impairing the EAAC1-mediated inward transport of Cysteine (Cys) and this can affect fetal brain development. In this study, we investigated if there is a role for the transulfuration pathway (TSP), a critical bio-synthetic point to supply Cys in E-induced dysregulation of GSH homeostasis. These studies utilized an in utero E binge model where the pregnant Sprague⁻Dawley (SD) rat dams received five doses of E at 3.5 g/kg by gastric intubation beginning embryonic day (ED) 17 until ED19 separated by 12 h. The postnatal day 7 (PN7) alcohol model employed an oral dosing of 4 g/kg body weight split into 2 feedings at 2 h interval and an iso-caloric and iso-volumic equivalent maltose-dextrin milk solution served as controls. The in vitro model consisted of cerebral cortical neuron cultures from embryonic day (ED) 16⁻17 fetus from SD rats and differentiated neurons from ED18 rat cerebral cortical neuroblasts. E concentrations were 4 mg/mL. E induced an accumulation of cystathionine in primary cortical neurons (PCNs), 2nd trimester equivalent in utero binge, and 3rd trimester equivalent PN7 model suggesting that breakdown of cystathionine, a required process for Cys supply is impaired. This was associated with a significant reduction in cystathionine γ-lyase (CSE) protein expression in PCN (p < 0.05) and in fetal cerebral cortex in utero (53%, p < 0.05) without a change in the expression of cystathionine β-synthase (CBS). Concomitantly, E decreased Cse mRNA expression in PCNs (by 32% within 6 h of exposure, p < 0.05) and in fetal brain (33%, p < 0.05). In parallel, knock down of CSE in differentiated rat cortical neuroblasts exaggerated the E-induced ROS, GSH loss with a pronounced caspase-3 activation and cell death. These studies illustrate the importance of TSP in CSE-related maintenance of GSH and the downstream events via Cys synthesis in neurons and fetal brain.
Collapse
Affiliation(s)
- Dhyanesh Patel
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.
| | - Marylatha Rathinam
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.
| | - Courtney Jarvis
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.
| | - Lenin Mahimainathan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.
| | - George Henderson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.
| | - Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.
| |
Collapse
|
39
|
Najm IM, Sarnat HB, Blümcke I. Review: The international consensus classification of Focal Cortical Dysplasia - a critical update 2018. Neuropathol Appl Neurobiol 2018; 44:18-31. [DOI: 10.1111/nan.12462] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022]
Affiliation(s)
- I. M. Najm
- Cleveland Clinic Epilepsy Centre; Cleveland OH USA
| | - H. B. Sarnat
- Faculty of Medicine; Departments of Paediatrics, Pathology (Neuropathology) and Clinical Neurosciences; University of Calgary; Calgary AB Canada
| | - I. Blümcke
- Department of Neuropathology; University Hospital; Erlangen Germany
| |
Collapse
|
40
|
Somatic mutations rather than viral infection classify focal cortical dysplasia type II as mTORopathy. Curr Opin Neurol 2018; 29:388-95. [PMID: 26840044 DOI: 10.1097/wco.0000000000000303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Genetic studies in focal cortical dysplasia type II (FCD II) provided ample evidence for somatic mutations in genes associated with the mammalian target of rapamycin (mTOR) pathway. Interestingly, the mTOR pathway can also be activated by the E6 oncogene of human papilloma viruses, and available data in FCD II remain controversial. We review and discuss the contradicting etiologies. RECENT FINDINGS The neuroembryologic basis of cortical development and timing of a somatic mutation occurring in proliferating neuroblasts can mechanistically link mTORopathies. When a somatic mutation occurs in proliferating neuroblasts at an early stage of their anticipated total number of 33 mitotic cell cycles, large hemispheric lesions will develop from their affected progeny. Somatic mutations occurring at later periods of neuroblast expansion will result in circumscribed and small FCD II. Recently published data did not support evidence for viral infection in FCD II. SUMMARY Genetic and histopathological data rather than viral infection classify FCD II into the spectrum of mTORopathies. Size and extent of the resulting cerebral lesion can be well explained by timing of somatic mutations during cortical development.
Collapse
|
41
|
Cerebellar networks and neuropathology of cerebellar developmental disorders. HANDBOOK OF CLINICAL NEUROLOGY 2018; 154:109-128. [PMID: 29903435 DOI: 10.1016/b978-0-444-63956-1.00007-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cerebellar system is a series of axonal projections and synaptic circuits as networks, similar to those of the limbic system and those subserving the propagation and spread of seizures. Three principal cerebellar networks are identified and cerebellar disease often affects components of the networks other than just the cerebellar cortex. Contemporary developmental neuropathology of the cerebellum is best considered in the context of alterations of developmental processes: embryonic segmentation and genetic gradients along the three axes of the neural tube, individual neuronal and glial cell differentiation, migration, synaptogenesis, and myelination. Precisely timed developmental processes may be delayed or precocious rhombencephalosynapsis and pontocerebellar hypoplasia exemplify opposite gradients in the horizontal axis. Chiari II malformation may be reconsidered as a disorder of segmentation rather than simply due to mechanical forces upon normally developing hindbrain structures. Cellular nodules in the roof of the fourth ventricle are heterotopia of histologically differentiated but architecturally disoriented and disorganized neurons and glial cells; they often are less mature immunocytochemically than similar cells in adjacent normal folia. Cell rests are nodules of undifferentiated neuroepithelial cells. Both are frequent in human fetuses and neonates. Axonal projections from heterotopia to adjacent cerebellar folia or nuclei are few or absent, hence these nodules are clinically silent despite neuronal differentiation.
Collapse
|
42
|
Zobel K, Choi SE, Minakova R, Gocyla M, Offenhäusser A. N-Cadherin modified lipid bilayers promote neural network formation and circuitry. SOFT MATTER 2017; 13:8096-8107. [PMID: 29085948 DOI: 10.1039/c7sm01214d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Neural adhesion, maturation, and the correct wiring of the brain to establish each neuron's intended connectivity are controlled by complex interactions of bioactive molecules such as ligands, growth factors, or enzymes. The correct pairing of adjacent neurons is thought to be highly regulated by ligand-mediated cell-cell adhesion proteins, which are known to induce signaling activities. We developed a new platform consisting of supported lipid bilayers incorporated with Fc-chimera synaptic proteins like ephrinA5 or N-cadherin. We extensively characterized their function employing a quartz crystal microbalance with dissipation (QCM-D), calcium imaging, and immunofluorescence analysis. Our biomimetic platform has been shown to promote neural cell adhesion and to improve neural maturation at day in vitro 7 (DIV7) as indicated by an elevated expression of synaptophysin.
Collapse
Affiliation(s)
- K Zobel
- Institute of Bioelectronics (ICS-8), Forschungszentrum Juelich, Wilhelm-Johnen Straße, 52425 Juelich, Germany.
| | | | | | | | | |
Collapse
|
43
|
Sarnat HB, Scantlebury MH. Novel Inflammatory Neuropathology in Immature Brain: (1) Fetal Tuberous Sclerosis, (2) Febrile Seizures, (3) α-B-crystallin, and (4) Role of Astrocytes. Semin Pediatr Neurol 2017; 24:152-160. [PMID: 29103422 DOI: 10.1016/j.spen.2017.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Though the term "inflammation" is traditionally defined as proliferation or infiltration of lymphatic cells of the lymphatic immune system and macrophages or as immunoreactive proteins including cytokines, interleukins and major histocompatibility complexes, recently recognized reactions to tissue injury also are inflammation, often occurring in the central nervous system in conditions where they previously were not anticipated and where they may play a role in both pathogenesis and repair. We highlight 4 such novel inflammatory conditions revealed by neuropathologic studies: (1) inflammatory markers and cells in the brain of human fetuses with tuberous sclerosis complex and perhaps other disorders of the mechanistic target of rapamycin genetic or metabolic pathway, (2) inflammatory markers in the brain related to febrile seizures of infancy and early childhood, (3) heat-shock protein upregulation in glial cells and neurons at sites of chronic epileptic foci, and (4) the emerging role of astrocytes in the presence of and participation in inflammation. Novel evidence shows that cerebral inflammation plays a role in some genetic diseases as early as midgestation and thus is not always acquired postnatally or in adult life.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Department of Pediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.
| | - Morris H Scantlebury
- Department of Pediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| |
Collapse
|
44
|
Laquerriere A, Maillard C, Cavallin M, Chapon F, Marguet F, Molin A, Sigaudy S, Blouet M, Benoist G, Fernandez C, Poirier K, Chelly J, Thomas S, Bahi-Buisson N. Neuropathological Hallmarks of Brain Malformations in Extreme Phenotypes Related to DYNC1H1 Mutations. J Neuropathol Exp Neurol 2017; 76:195-205. [PMID: 28395088 DOI: 10.1093/jnen/nlw124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dyneins play a critical role in a wide variety of cellular functions such as the movement of organelles and numerous aspects of mitosis, making it central player in neocortical neurogenesis and migration. Recently, cytoplasmic dynein-1, heavy chain-1 (DYNC1H1) mutations have been found to cause a wide spectrum of brain cortical malformations. We report on the detailed neuropathological features of brain lesions from 2 fetuses aged 36 and 22 weeks of gestation (WG), respectively, carrying de novo DYNC1H1 mutations, p.Arg2720Lys and p.Val3951Ala and presenting the most severe phenotype reported to date. Analysis using the Dictyostelium discoideum dynein motor crystal structure showed that the mutations are both predicted to have deleterious consequences on the function of the motor domain. Both fetuses showed a similar macroscopic and histological brain malformative complex associating bilateral fronto-parietal polymicrogyria (PMG), dysgenesis of the corpus callosum and of the cortico-spinal tracts, along with brainstem and cerebellar abnormalities. Both exhibited extremely severe disrupted cortical lamination. Immunohistochemical studies provided the evidence for defects in cell proliferation and postmitotic neuroblast ability to exit from the subventricular zone resulting in a failure of radial migration toward the cortical plate, thus providing new insights for the understanding of the pathophysiology in these cortical malformations.
Collapse
Affiliation(s)
- Annie Laquerriere
- Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Normandie University, Rouen University Hospital, NeoVasc Team, UNIROUEN, Inserm U1245, Rouen, France
| | - Camille Maillard
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris and INSERM UMR-1163, Embryology and Genetics of Congenital Malformations, Paris France
| | - Mara Cavallin
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris and INSERM UMR-1163, Embryology and Genetics of Congenital Malformations, Paris France.,Pediatric Neurology, Necker Enfants Malades University Paris Hospital, APHP, Paris, France
| | | | - Florent Marguet
- Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Normandie University, Rouen University Hospital, NeoVasc Team, UNIROUEN, Inserm U1245, Rouen, France
| | - Arnaud Molin
- Service de Génétique Pôle Biologie et Pharmacie Rouen University Hôpital, France
| | - Sabine Sigaudy
- Clinical Genetics, Hôpital de La Timone, APHM, Marseille University Hospital, Marseille France
| | - Marie Blouet
- Department of Radiology, Caen University Hospital, Caen France
| | - Guillaume Benoist
- Department of Obstetrics and Gynaecology, Caen University Hospital, Caen France
| | - Carla Fernandez
- Department of Pathology and Neuropathology, La Timone University Hospital, Marseille, France
| | - Karine Poirier
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Denis Diderot School of Medicine, Sorbonne-Paris Cité University, Paris, France
| | - Jamel Chelly
- IGBMC, INSERM U964, CNRS UMR 7104, Université de Strasbourg, Illkirch, France.,Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Sophie Thomas
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris and INSERM UMR-1163, Embryology and Genetics of Congenital Malformations, Paris France
| | - Nadia Bahi-Buisson
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris and INSERM UMR-1163, Embryology and Genetics of Congenital Malformations, Paris France.,Pediatric Neurology, Necker Enfants Malades University Paris Hospital, APHP, Paris, France
| |
Collapse
|
45
|
Kreiner G, Rafa-Zabłocka K, Chmielarz P, Bagińska M, Nalepa I. Lack of riluzole efficacy in the progression of the neurodegenerative phenotype in a new conditional mouse model of striatal degeneration. PeerJ 2017; 5:e3240. [PMID: 28462043 PMCID: PMC5410142 DOI: 10.7717/peerj.3240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/28/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is a rare familial autosomal dominant neurodegenerative disorder characterized by progressive degeneration of medium spiny neurons (MSNs) located in the striatum. Currently available treatments of HD are only limited to alleviating symptoms; therefore, high expectations for an effective therapy are associated with potential replacement of lost neurons through stimulation of postnatal neurogenesis. One of the drugs of potential interest for the treatment of HD is riluzole, which may act as a positive modulator of adult neurogenesis, promoting replacement of damaged MSNs. The aim of this study was to evaluate the effects of chronic riluzole treatment on a novel HD-like transgenic mouse model, based on the genetic ablation of the transcription factor TIF-IA. This model is characterized by selective and progressive degeneration of MSNs. METHODS Selective ablation of TIF-IA in MSNs (TIF-IAD1RCre mice) was achieved by Cre-based recombination driven by the dopamine 1 receptor (D1R) promoter in the C57Bl/6N mouse strain. Riluzole was administered for 14 consecutive days (5 mg/kg, i.p.; 1× daily) starting at six weeks of age. Behavioral analysis included a motor coordination test performed on 13-week-old animals on an accelerated rotarod (4-40 r.p.m.; 5 min). To visualize the potential effects of riluzole treatment, the striata of the animals were stained by immunohistochemistry (IHC) and/or immunofluorescence (IF) with Ki67 (marker of proliferating cells), neuronal markers (NeuN, MAP2, DCX), and markers associated with neurodegeneration (GFAP, 8OHdG, FluoroJade C). Additionally, the morphology of dendritic spines of neurons was assessed by a commercially available FD Rapid Golgi Stain™ Kit. RESULTS A comparative analysis of IHC staining patterns with chosen markers for the neurodegeneration process in MSNs did not show an effect of riluzole on delaying the progression of MSN cell death despite an observed enhancement of cell proliferation as visualized by the Ki67 marker. A lack of a riluzole effect was also reflected by the behavioral phenotype associated with MSN degeneration. Moreover, the analysis of dendritic spine morphology did not show differences between mutant and control animals. DISCUSSION Despite the observed increase in newborn cells in the subventricular zone (SVZ) after riluzole administration, our study did not show any differences between riluzole-treated and non-treated mutants, revealing a similar extent of the neurodegenerative phenotype evaluated in 13-week-old TIF-IAD1RCre animals. This could be due to either the treatment paradigm (relatively low dose of riluzole used for this study) or the possibility that the effects were simply too weak to have any functional meaning. Nevertheless, this study is in line with others that question the effectiveness of riluzole in animal models and raise concerns about the utility of this drug due to its rather modest clinical efficacy.
Collapse
Affiliation(s)
- Grzegorz Kreiner
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Katarzyna Rafa-Zabłocka
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Piotr Chmielarz
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Monika Bagińska
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Irena Nalepa
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| |
Collapse
|
46
|
Abstract
Olfactory axons project from nasal epithelium to the primitive telencephalon before olfactory bulbs form. Olfactory bulb neurons do not differentiate in situ but arrive via the rostral migratory stream. Synaptic glomeruli and concentric laminar architecture are unlike other cortices. Fetal olfactory maturation of neuronal differentiation, synaptogenesis, and myelination remains incomplete at term and have a protracted course of postnatal development. The olfactory ventricular recess involutes postnatally but dilates in congenital hydrocephalus. Olfactory bulb, tract and epithelium are repositories of progenitor stem cells in fetal and adult life. Diverse malformations of the olfactory bulb can be diagnosed by clinical examination, imaging, and neuropathologically. Cellular markers of neuronal differentiation and synaptogenesis demonstrate immaturity of the olfactory system at birth, previously believed by histology alone to occur early in fetal life. Immaturity does not preclude function.
Collapse
Affiliation(s)
- Harvey B Sarnat
- 1 Department of Paediatrics, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.,2 Department of Pathology and Laboratory Medicine (Neuropathology), University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.,3 Department of Clinical Neurosciences, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Laura Flores-Sarnat
- 1 Department of Paediatrics, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.,3 Department of Clinical Neurosciences, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| |
Collapse
|
47
|
Abstract
Epilepsy is one of the most common neurologic disorders, affecting about 50 million people worldwide. The disease is characterized by recurrent seizures, which are due to aberrant neuronal networks resulting in synchronous discharges. The term epilepsy encompasses a large spectrum of syndromes and diseases with different etiopathogenesis. The recent development of imaging and epilepsy surgery techniques is now enabling the identification of structural abnormalities that are part of the epileptic network, and the removal of these lesions may result in control of seizures. Access of this clinically well-characterized neurosurgical material has provided neuropathologists with the opportunity to study a variety of structural brain abnormalities associated with epilepsy, by combining traditional routine histopathologic methods with molecular genetics and functional analysis of the resected tissue. This approach has contributed greatly to a better diagnosis and classification of these structural lesions, and has provided important new insights into their pathogenesis and epileptogenesis. The present chapter provides a detailed description of the large spectrum of histopathologic findings encountered in epilepsy surgery patients, addressing in particular the nonneoplastic pathologies, including hippocampal sclerosis, malformations of cortical development, Sturge-Weber syndrome, and Rasmussen encephalitis, and reviews current knowledge regarding the underlying molecular pathomechanisms and cellular mechanisms mediating hyperexcitability.
Collapse
Affiliation(s)
- Eleonora Aronica
- Department of Neuropathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland, the Netherlands.
| | - Angelika Mühlebner
- Department of Neuropathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
48
|
ALS disrupts spinal motor neuron maturation and aging pathways within gene co-expression networks. Nat Neurosci 2016; 19:1256-67. [PMID: 27428653 PMCID: PMC5003654 DOI: 10.1038/nn.4345] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/17/2016] [Indexed: 02/07/2023]
Abstract
Modeling Amyotrophic Lateral Sclerosis (ALS) with human induced pluripotent stem cells (iPSCs) aims to reenact embryogenesis, maturation, and aging of spinal motor neurons (spMNs) in vitro. As the maturity of spMNs grown in vitro compared to spMNs in vivo remains largely unaddressed, it is unclear to what extent this in vitro system captures critical aspects of spMN development and molecular signatures associated with ALS. Here, we compared transcriptomes among iPSC-derived spMNs, fetal, and adult spinal tissues. This approach produced a maturation scale revealing that iPSC-derived spMNs were more similar to fetal spinal tissue than to adult spMNs. Additionally, we resolved gene networks and pathways associated with spMN maturation and aging. These networks enriched for pathogenic familial ALS genetic variants and were disrupted in sporadic ALS spMNs. Altogether, our findings suggest that developing strategies to further mature and age iPSC-derived spMNs will provide more effective iPSC models of ALS pathology.
Collapse
|
49
|
Sarnat HB, Flores-Sarnat L. Synaptogenesis and Myelination in the Nucleus/Tractus Solitarius: Potential Role in Apnea of Prematurity, Congenital Central Hypoventilation, and Sudden Infant Death Syndrome. J Child Neurol 2016; 31:722-32. [PMID: 26661483 DOI: 10.1177/0883073815615227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/26/2015] [Indexed: 12/14/2022]
Abstract
Fetuses as early as 15 weeks' gestation exhibit rhythmical respiratory movements shown by real-time ultrasonography. The nucleus/tractus solitarius is the principal brainstem respiratory center; other medullary nuclei also participate. The purpose was to determine temporal maturation of synaptogenesis. Delayed synaptic maturation may explain neurogenic apnea or hypoventilation of prematurity and some cases of sudden infant death syndrome. Sections of medulla oblongata were studied from 30 human fetal and neonatal brains 9 to 41 weeks' gestation. Synaptophysin demonstrated the immunocytochemical sequence of synaptogenesis. Other neuronal markers and myelin stain also were applied. The nucleus/tractus solitarius was similarly studied in fetuses with chromosomopathies, metabolic encephalopathies, and brain malformations. Synapse formation in the nucleus solitarius begins at about 12 weeks' gestation and matures by 15 weeks; myelination initiated at 33 weeks. Synaptogenesis was delayed in 3 fetuses with different conditions, but was not specific for only nucleus solitarius. Delayed synaptogenesis or myelination in the nucleus solitarius may play a role in neonatal hypoventilation, especially in preterm infants and in some sudden infant death syndrome cases.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics, Pathology (Neuropathology) and Clinical Neurosciences, University of Calgary and Alberta Children's Hospital Research Institute, Calgary Alberta, Canada
| | - Laura Flores-Sarnat
- Departments of Paediatrics, Pathology (Neuropathology) and Clinical Neurosciences, University of Calgary and Alberta Children's Hospital Research Institute, Calgary Alberta, Canada
| |
Collapse
|
50
|
Sarnat HB, Yu W. Maturation and Dysgenesis of the Human Olfactory Bulb. Brain Pathol 2016; 26:301-18. [PMID: 26096058 PMCID: PMC8028954 DOI: 10.1111/bpa.12275] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/09/2015] [Indexed: 12/22/2022] Open
Abstract
The olfactory bulb with its unique architecture was studied for neuronal maturation in human fetuses. Neuroblasts stream into the olfactory bulb from the rostral telencephalon and secondarily migrate radially. The transitory olfactory ventricular recess regresses postnatally. Olfactory is the only sensory system without thalamic projections but incorporates intrinsic thalamic equivalents. The bulb is a repository of progenitor cells. Maturation of the bulb and tract was studied in 18 normal human fetuses of 16-41 weeks gestation; mid-gestational twins with hydrocephalus; 7 arrhinencephaly/holoprosencephaly; 2 olfactory dysgeneses. Multiple immunoreactivities were performed. Synaptophysin around mitral neurons, in a few synaptic glomeruli and concentric lamination of the outer granular layer, was seen at 16 weeks. Outer granular neurons exhibited NeuN at 16 weeks, only 2/3 were reactive at term. Concentric alternating sheets of granular neurons and their dendrodendritic synapses are seen during maturation. Calretinin reactivity is seen in neurons and neurites, primary olfactory nerve axons, periglomerular cells and neuroepithelial cells surrounding the ventricular recess; reactivity occurs later in synaptic glomeruli than with synaptophysin; not all glomeruli are strongly reactive even at term. Nestin- and vimentin-reactive bipolar progenitor cells were demonstrated at all ages and extend into the olfactory tract. Myelin is demonstrated by Luxol fast blue (LFB) only postnatally. In hydrocephalus, the olfactory recess is dilated. Mitral cell dispersion, disrupted glomeruli, heterotopia and maturational delay are seen in some dysgeneses. Malformations exhibit unique findings. Fusion of hypoplastic bulbs can occur. Abnormal architecture is seen in hemimegalencephaly. More documentation of olfactory dysgenesis is needed in other major brain malformations.
Collapse
Affiliation(s)
- Harvey B. Sarnat
- Department of PaediatricsUniversity of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
- Department of Pathology and Laboratory Medicine (Neuropathology)University of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
- Department of Clinical NeurosciencesUniversity of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
| | - Weiming Yu
- Department of PaediatricsUniversity of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
- Department of Pathology and Laboratory Medicine (Paediatric Pathology)University of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
| |
Collapse
|