1
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Ben Dhaou C, Terrié E, Déliot N, Harnois T, Cousin L, Arnault P, Constantin B, Moyse E, Coronas V. Neural stem cell self-renewal stimulation by store-operated calcium entries in adult mouse area postrema: influence of leptin. Front Cell Neurosci 2023; 17:1200360. [PMID: 37361995 PMCID: PMC10287973 DOI: 10.3389/fncel.2023.1200360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/17/2023] [Indexed: 06/28/2023] Open
Abstract
Neural stem cells (NSCs) persist in specific brain germinative niches and sustain neurogenesis throughout life in adult mammals. In addition to the two major stem cell niches in the subventricular zone and the hippocampal dentate gyrus, the area postrema located in the brainstem has been identified as a neurogenic zone as well. NSCs are regulated by signals from the microenvironment that adjust stem cell response to the needs of the organism. Evidence accumulated over the past decade indicates that Ca2+ channels play pivotal functions in NSC maintenance. In this study, we explored in area postrema NSCs the presence and roles of a subset of Ca2+ channels, the store-operated Ca2+ channels (SOCs) that have the capacity to transduce extracellular signals into Ca2+ signals. Our data show that NSCs derived from the area postrema express TRPC1 and Orai1, known to form SOCs, as well as their activator STIM1. Ca2+ imaging indicated that NSCs exhibit store-operated Ca2+ entries (SOCEs). Pharmacological blockade of SOCEs with SKF-96365, YM-58483 (also known as BTP2) or GSK-7975A resulted in decreased NSC proliferation and self-renewal, indicating a major role for SOCs in maintaining NSC activity within the area postrema. Furthermore, our results show that leptin, an adipose tissue-derived hormone whose ability to control energy homeostasis is dependent on the area postrema, decreased SOCEs and reduced self-renewal of NSCs in the area postrema. As aberrant SOC function has been linked to an increasing number of diseases, including brain disorders, our study opens new perspectives for NSCs in brain pathophysiology.
Collapse
Affiliation(s)
- Cyrine Ben Dhaou
- University of Tours, INRAe Centre Val-de-Loire UMR-85, CNRS UMR-1247, Physiologie de la Reproduction et Comportements, Nouzilly, France
| | - Elodie Terrié
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Nadine Déliot
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Thomas Harnois
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Laetitia Cousin
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Patricia Arnault
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Bruno Constantin
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Emmanuel Moyse
- University of Tours, INRAe Centre Val-de-Loire UMR-85, CNRS UMR-1247, Physiologie de la Reproduction et Comportements, Nouzilly, France
| | - Valérie Coronas
- 4CS, Laboratory Channels and Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| |
Collapse
|
3
|
Iwata K, Ferdousi F, Arai Y, Isoda H. Interactions between Major Bioactive Polyphenols of Sugarcane Top: Effects on Human Neural Stem Cell Differentiation and Astrocytic Maturation. Int J Mol Sci 2022; 23:ijms232315120. [PMID: 36499441 PMCID: PMC9738893 DOI: 10.3390/ijms232315120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Sugarcane (Saccharum officinarum L.) is a tropical plant grown for sugar production. We recently showed that sugarcane top (ST) ameliorates cognitive decline in a mouse model of accelerated aging via promoting neuronal differentiation and neuronal energy metabolism and extending the length of the astrocytic process in vitro. Since the crude extract consists of multicomponent mixtures, it is crucial to identify bioactive compounds of interest and the affected molecular targets. In the present study, we investigated the bioactivities of major polyphenols of ST, namely 3-O-caffeoylquinic acid (3CQA), 5-O-caffeoylquinic acid (5CQA), 3-O-feruloylquinic acid (3FQA), and Isoorientin (ISO), in human fetal neural stem cells (hNSCs)- an in vitro model system for studying neural development. We found that multiple polyphenols of ST contributed synergistically to stimulate neuronal differentiation of hNSCs and induce mitochondrial activity in immature astrocytes. Mono-CQAs (3CQA and 5CQA) regulated the expression of cyclins related to G1 cell cycle arrest, whereas ISO regulated basic helix-loop-helix transcription factors related to cell fate determination. Additionally, mono-CQAs activated p38 and ISO inactivated GSK3β. In hNSC-derived immature astrocytes, the compounds upregulated mRNA expression of PGC-1α, a master regulator of astrocytic mitochondrial biogenesis. Altogether, our findings suggest that synergistic interactions between major polyphenols of ST contribute to its potential for neuronal differentiation and astrocytic maturation.
Collapse
Affiliation(s)
- Kengo Iwata
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8572, Japan
- Nipoo Co., Ltd., Osaka 574-0062, Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8572, Japan
- AIST—University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba 305-8572, Japan
| | | | - Hiroko Isoda
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8572, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8572, Japan
- AIST—University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba 305-8572, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
- Correspondence: ; Tel.: +81-29-853-5775
| |
Collapse
|
4
|
Laaksonen K, Ward NS. Biomarkers of plasticity for stroke recovery. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:287-298. [PMID: 35034742 DOI: 10.1016/b978-0-12-819410-2.00033-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Stroke is the commonest cause of physical disability in the world. Our understanding of the biologic mechanisms involved in recovery and repair has advanced to the point that therapeutic opportunities to promote recovery through manipulation of post-stroke plasticity have never been greater. This work has almost exclusively been carried out in rodent models of stroke with little translation into human studies. The challenge ahead is to develop a mechanistic understanding of recovery from stroke in humans. Advances in neuroimaging techniques can now provide the appropriate intermediate level of description to bridge the gap between a molecular and cellular account of recovery and a behavioral one. Clinical trials can then be designed in a stratified manner taking into account when an intervention should be delivered and who is most likely to benefit. This approach is most likely to lead to the step-change in how restorative therapeutic strategies are delivered in human stroke patients.
Collapse
Affiliation(s)
- Kristina Laaksonen
- Department of Neurology, Helsinki University Hospital, and Clinical Neurosciences, Neurology, University of Helsinki, Helsinki, Finland
| | - Nick S Ward
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom.
| |
Collapse
|
5
|
Buzoianu-Anguiano V, Torres-Llacsa M, Doncel-Pérez E. Role of Aldynoglia Cells in Neuroinflammatory and Neuroimmune Responses after Spinal Cord Injury. Cells 2021; 10:2783. [PMID: 34685763 PMCID: PMC8534338 DOI: 10.3390/cells10102783] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Aldynoglia are growth-promoting cells with a morphology similar to radial glia and share properties and markers with astrocytes and Schwann cells. They are distributed in several locations throughout the adult central nervous system, where the cells of the aldynoglia interact and respond to the signals of the immune cells. After spinal cord injury (SCI), the functions of resident aldynoglia, identified as ependymocytes, tanycytes, and ependymal stem cells (EpSCs) of the spinal cord are crucial for the regeneration of spinal neural tissue. These glial cells facilitate axonal regrowth and remyelination of injured axons. Here, we review the influence of M1 or M2 macrophage/microglia subpopulations on the fate of EpSCs during neuroinflammation and immune responses in the acute, subacute, and chronic phases after SCI.
Collapse
Affiliation(s)
| | - Mabel Torres-Llacsa
- Servicio de Radiología, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain;
| | - Ernesto Doncel-Pérez
- Grupo de Química Neuro-Regenerativa, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain;
| |
Collapse
|
6
|
El Nahas N, Elbokl AM, Abd Eldayem EH, Roushdy TM, Amin RM, Helmy SM, Akl AZ, Ashour AA, Samy S, Amgad A, Emara TH, Nowara M, Kenawy FF. Navigated perilesional transcranial magnetic stimulation can improve post-stroke visual field defect: A double-blind sham-controlled study. Restor Neurol Neurosci 2021; 39:199-207. [PMID: 34024791 DOI: 10.3233/rnn-211181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Visual field defects (VFD) usually do not show improvement beyond 12 weeks from their onset. It has been shown that repetitive presentation of a stimulus to areas of residual vision in cases of visual field defect can improve vision. The counterpart of these areas in the brain are the partially damaged brain regions at the perilesional areas where plasticity can be enhanced. OBJECTIVE We aimed to study the effect of navigated repetitive transcranial magnetic stimulation (rTMS) applied to perilesional areas on the recovery of patients with cortical VFD. METHODS Thirty-two patients with cortical VFD secondary to stroke of more than 3 months duration received 16 sessions of either active or sham high frequency navigated perilesional rTMS. Automated perimetry and visual functioning questionnaire (VFQ-25) were performed at baseline and after completion of the sessions. RESULTS The active group showed significant improvement after intervention, compared to the sham group, in both mean deviation (MD), visual field index (VFI) and in the VFQ-25 scores. CONCLUSIONS Navigated rTMS is a new treatment option for post-stroke VFD as it can selectively stimulate areas of residual vision around the infarcted tissue, improving the threshold of visual stimulus detection which could be used alone or in combination with existing therapies.
Collapse
Affiliation(s)
- Nevine El Nahas
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed M Elbokl
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman Hamid Abd Eldayem
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tamer M Roushdy
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Randa M Amin
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Shahinaz M Helmy
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Zaki Akl
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Aya Ahmed Ashour
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Shady Samy
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alaa Amgad
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tamer H Emara
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Fatma Fathalla Kenawy
- Ain Shams Neuromodulation Research Lab, Neurology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
7
|
Nambu Y, Ohira K, Morita M, Yasumoto H, Kurganov E, Miyata S. Effects of leptin on proliferation of astrocyte- and tanycyte-like neural stem cells in the adult mouse medulla oblongata. Neurosci Res 2021; 173:44-53. [PMID: 34058263 DOI: 10.1016/j.neures.2021.05.012] [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: 03/08/2021] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Astrocyte- and tanycyte-like neural stem cells (NSCs) were recently detected in the area postrema (AP) and central canal (CC) of the adult medulla oblongata, respectively. The present study aimed to examine dynamical behaviors of the astrocyte- and tanycyte-like NSCs of the mouse medulla oblongata to leptin. The neurosphere assay identified astrocytes in the AP and tanycytes in the CC as NSCs based on their self-renewing neurospherogenic potential. Both NSCs in neurosphere cultures were multipotent cells that generate astrocytes, oligodendrocytes, and neurons. Astrocyte-like NSCs actively proliferated and tanycyte-like NSCs were quiescent under physiologically-relevant in vivo conditions. Chronic leptin treatment promoted proliferation of astrocyte-like NSCs in the AP both in vitro and in vivo. Leptin receptors were expressed in astrocyte-like, but not tanycyte-like NSCs. Food deprivation significantly diminished proliferation of astrocyte-like NSCs. Therefore, the present study indicates that proliferation of astrocyte-like, but not tanycyte-like NSCs is regulated by nutritional conditions.
Collapse
Affiliation(s)
- Yuri Nambu
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Koji Ohira
- Laboratory of Nutritional Brain Science, Department of Food Science and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan
| | - Hiroki Yasumoto
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Erkin Kurganov
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan.
| |
Collapse
|
8
|
Nambu Y, Horie K, Kurganov E, Miyata S. Chronic running and a corticosterone treatment attenuate astrocyte-like neural stem cell proliferation in the area postrema of the adult mouse brain. Neurosci Lett 2021; 748:135732. [PMID: 33592302 DOI: 10.1016/j.neulet.2021.135732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 01/25/2023]
Abstract
The discovery of neural stem cells (NSCs) in the adult mammalian brain has provided insights into an extra level of brain plasticity. The proliferation and differentiation of NSCs is modulated by various physiological, pathological, and pharmacological stimuli. NSCs were recently detected in the medulla oblongata of adult rodents and humans; however, their functional significance currently remains unknown. In the present study, we examined the effects of chronic wheel-running and a corticosterone (CORT) treatment on the proliferation of astrocyte-like NSCs in the area postrema (AP) and dentate gyrus (DG). Chronic running significantly decreased the number of bromodeoxyuridine (BrdU)-labeled astrocyte-like NSCs in the AP of adult mice, but markedly increased that of BrdU+ NSCs/neural progenitor cells in the DG. The chronic CORT treatment markedly reduced the number of BrdU+ astrocyte-like NSCs in the AP, but not in the DG. These results demonstrate that the proliferation of astrocyte-like NSCs in the medulla oblongata is decreased by chronic running and a CORT treatment.
Collapse
Affiliation(s)
- Yuri Nambu
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Kohei Horie
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Erkin Kurganov
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
9
|
Bernardo-Castro S, Sousa JA, Brás A, Cecília C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, Santana I, Sargento-Freitas J. Pathophysiology of Blood-Brain Barrier Permeability Throughout the Different Stages of Ischemic Stroke and Its Implication on Hemorrhagic Transformation and Recovery. Front Neurol 2020; 11:594672. [PMID: 33362697 PMCID: PMC7756029 DOI: 10.3389/fneur.2020.594672] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/09/2020] [Indexed: 12/25/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic interface responsible for maintaining the central nervous system homeostasis. Its unique characteristics allow protecting the brain from unwanted compounds, but its impairment is involved in a vast number of pathological conditions. Disruption of the BBB and increase in its permeability are key in the development of several neurological diseases and have been extensively studied in stroke. Ischemic stroke is the most prevalent type of stroke and is characterized by a myriad of pathological events triggered by an arterial occlusion that can eventually lead to fatal outcomes such as hemorrhagic transformation (HT). BBB permeability seems to follow a multiphasic pattern throughout the different stroke stages that have been associated with distinct biological substrates. In the hyperacute stage, sudden hypoxia damages the BBB, leading to cytotoxic edema and increased permeability; in the acute stage, the neuroinflammatory response aggravates the BBB injury, leading to higher permeability and a consequent risk of HT that can be motivated by reperfusion therapy; in the subacute stage (1-3 weeks), repair mechanisms take place, especially neoangiogenesis. Immature vessels show leaky BBB, but this permeability has been associated with improved clinical recovery. In the chronic stage (>6 weeks), an increase of BBB restoration factors leads the barrier to start decreasing its permeability. Nonetheless, permeability will persist to some degree several weeks after injury. Understanding the mechanisms behind BBB dysregulation and HT pathophysiology could potentially help guide acute stroke care decisions and the development of new therapeutic targets; however, effective translation into clinical practice is still lacking. In this review, we will address the different pathological and physiological repair mechanisms involved in BBB permeability through the different stages of ischemic stroke and their role in the development of HT and stroke recovery.
Collapse
Affiliation(s)
| | - João André Sousa
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Ana Brás
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Carla Cecília
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Bruno Rodrigues
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Luciano Almendra
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Cristina Machado
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Gustavo Santo
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Fernando Silva
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
10
|
Zhang W, Zhu L, An C, Wang R, Yang L, Yu W, Li P, Gao Y. The blood brain barrier in cerebral ischemic injury – Disruption and repair. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2019.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
11
|
Neural stem cell phenotype of tanycyte-like ependymal cells in the circumventricular organs and central canal of adult mouse brain. Sci Rep 2020; 10:2826. [PMID: 32071335 PMCID: PMC7029029 DOI: 10.1038/s41598-020-59629-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/03/2020] [Indexed: 12/26/2022] Open
Abstract
Tanycyte is a subtype of ependymal cells which extend long radial processes to brain parenchyma. The present study showed that tanycyte-like ependymal cells in the organum vasculosum of the lamina terminalis, subfornical organ and central canal (CC) expressed neural stem cell (NSC) marker nestin, glial fibrillar acidic protein and sex determining region Y. Proliferation of these tanycyte-like ependymal cells was promoted by continuous intracerebroventricular infusion of fibroblast growth factor-2 and epidermal growth factor. Tanycytes-like ependymal cells in the CC are able to form self-renewing neurospheres and give rise mostly to new astrocytes and oligodendrocytes. Collagenase-induced small medullary hemorrhage increased proliferation of tanycyte-like ependymal cells in the CC. These results demonstrate that these tanycyte-like ependymal cells of the adult mouse brain are NSCs and suggest that they serve as a source for providing new neuronal lineage cells upon brain damage in the medulla oblongata.
Collapse
|
12
|
Abstract
The blood-brain barrier (BBB) protects the vertebrate central nervous system from harmful blood-borne, endogenous and exogenous substances to ensure proper neuronal function. The BBB describes a function that is established by endothelial cells of CNS vessels in conjunction with pericytes, astrocytes, neurons and microglia, together forming the neurovascular unit (NVU). Endothelial barrier function is crucially induced and maintained by the Wnt/β-catenin pathway and requires intact NVU for proper functionality. The BBB and the NVU are characterized by a specialized assortment of molecular specializations, providing the basis for tightening, transport and immune response functionality.The present chapter introduces state-of-the-art knowledge of BBB structure and function and highlights current research topics, aiming to understanding in more depth the cellular and molecular interactions at the NVU, determining functionality of the BBB in health and disease, and providing novel potential targets for therapeutic BBB modulation. Moreover, we highlight recent advances in understanding BBB and NVU heterogeneity within the CNS as well as their contribution to CNS physiology, such as neurovascular coupling, and pathophysiology, is discussed. Finally, we give an outlook onto new avenues of BBB research.
Collapse
Affiliation(s)
- Fabienne Benz
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Excellence Cluster Cardio Pulmonary System (CPI), Partner Site Frankfurt, Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.
| |
Collapse
|
13
|
Regenhardt RW, Takase H, Lo EH, Lin DJ. Translating concepts of neural repair after stroke: Structural and functional targets for recovery. Restor Neurol Neurosci 2020; 38:67-92. [PMID: 31929129 PMCID: PMC7442117 DOI: 10.3233/rnn-190978] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stroke is among the most common causes of adult disability worldwide, and its disease burden is shifting towards that of a long-term condition. Therefore, the development of approaches to enhance recovery and augment neural repair after stroke will be critical. Recovery after stroke involves complex interrelated systems of neural repair. There are changes in both structure (at the molecular, cellular, and tissue levels) and function (in terms of excitability, cortical maps, and networks) that occur spontaneously within the brain. Several approaches to augment neural repair through enhancing these changes are under study. These include identifying novel drug targets, implementing rehabilitation strategies, and developing new neurotechnologies. Each of these approaches has its own array of different proposed mechanisms. Current investigation has emphasized both cellular and circuit-based targets in both gray and white matter, including axon sprouting, dendritic branching, neurogenesis, axon preservation, remyelination, blood brain barrier integrity, blockade of extracellular inhibitory signals, alteration of excitability, and promotion of new brain cortical maps and networks. Herein, we review for clinicians recovery after stroke, basic elements of spontaneous neural repair, and ongoing work to augment neural repair. Future study requires alignment of basic, translational, and clinical research. The field continues to grow while becoming more clearly defined. As thrombolysis changed stroke care in the 1990 s and thrombectomy in the 2010 s, the augmentation of neural repair and recovery after stroke may revolutionize care for these patients in the coming decade.
Collapse
Affiliation(s)
- Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Hajime Takase
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Eng H Lo
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - David J Lin
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| |
Collapse
|
14
|
Cirillo C, Brihmat N, Castel-Lacanal E, Le Friec A, Barbieux-Guillot M, Raposo N, Pariente J, Viguier A, Simonetta-Moreau M, Albucher JF, Olivot JM, Desmoulin F, Marque P, Chollet F, Loubinoux I. Post-stroke remodeling processes in animal models and humans. J Cereb Blood Flow Metab 2020; 40:3-22. [PMID: 31645178 PMCID: PMC6928555 DOI: 10.1177/0271678x19882788] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
After cerebral ischemia, events like neural plasticity and tissue reorganization intervene in lesioned and non-lesioned areas of the brain. These processes are tightly related to functional improvement and successful rehabilitation in patients. Plastic remodeling in the brain is associated with limited spontaneous functional recovery in patients. Improvement depends on the initial deficit, size, nature and localization of the infarction, together with the sex and age of the patient, all of them affecting the favorable outcome of reorganization and repair of damaged areas. A better understanding of cerebral plasticity is pivotal to design effective therapeutic strategies. Experimental models and clinical studies have fueled the current understanding of the cellular and molecular processes responsible for plastic remodeling. In this review, we describe the known mechanisms, in patients and animal models, underlying cerebral reorganization and contributing to functional recovery after ischemic stroke. We also discuss the manipulations and therapies that can stimulate neural plasticity. We finally explore a new topic in the field of ischemic stroke pathophysiology, namely the brain-gut axis.
Collapse
Affiliation(s)
- Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Nabila Brihmat
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Evelyne Castel-Lacanal
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | | | - Nicolas Raposo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jérémie Pariente
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alain Viguier
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Marion Simonetta-Moreau
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-François Albucher
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-Marc Olivot
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Philippe Marque
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - François Chollet
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| |
Collapse
|
15
|
Morita-Takemura S, Wanaka A. Blood-to-brain communication in the hypothalamus for energy intake regulation. Neurochem Int 2019; 128:135-142. [DOI: 10.1016/j.neuint.2019.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 01/03/2023]
|
16
|
Shiota Y, Nagai A, Sheikh AM, Mitaki S, Mishima S, Yano S, Haque MA, Kobayashi S, Yamaguchi S. Transplantation of a bone marrow mesenchymal stem cell line increases neuronal progenitor cell migration in a cerebral ischemia animal model. Sci Rep 2018; 8:14951. [PMID: 30297706 PMCID: PMC6175868 DOI: 10.1038/s41598-018-33030-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation is demonstrated to improve functional and pathological recovery in cerebral ischemia. To understand the underlying mechanism, we transplanted a MSC line (B10) in a rat middle cerebral artery occlusion (MCAO) model and checked the proliferation and migration of neuronal progenitor cells (NPCs). B10 transplantation increased NPCs in the subventricular zone and their migration towards the lesion area at an earlier time. Fourteen days after MCAO, some NPCs were differentiated to neurons and astrocytes. Although B10 transplantation increased total number of both astrocytes and neurons, it only increased the differentiation of NPC to astrocyte. The mRNA of polysialylation enzyme ST8SiaIV and a chemokine SDF-1 were persistently increased in B10-transplanted groups. SDF-1-positive cell number was increased in the core and penumbra area, which was expressed in macrophage/microglia and transplanted B10 cells at 3 days after MCAO. Furthermore, SDF-1 mRNA expression in cell culture was high in B10 compared to a microglia (HMO) or a neuronal (A1) cell line. B10 culture supernatant increased in vitro A1 cell migration, which was significantly inhibited by siRNA-mediated SDF-1 silencing in B10. Thus, our results suggested that MSC transplantation increased endogenous NPC migration in cerebral ischemic condition by increasing chemokine and polysialylation enzyme expression, which could be helpful for the restorative management of cerebral ischemia.
Collapse
Affiliation(s)
- Yuri Shiota
- Clinical Laboratory Division, Shimane University Hospital, 89-1 Enya Cho, Izumo, 693-8501, Japan
| | - Atsushi Nagai
- Clinical Laboratory Division, Shimane University Hospital, 89-1 Enya Cho, Izumo, 693-8501, Japan. .,Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo, 693-8501, Japan.
| | - Abdullah Md Sheikh
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo, 693-8501, Japan
| | - Shingo Mitaki
- Department of Internal Medicine III, Shimane University School of Medicine, 89-1 Enya Cho, Izumo, 693-8501, Japan
| | - Seiji Mishima
- Clinical Laboratory Division, Shimane University Hospital, 89-1 Enya Cho, Izumo, 693-8501, Japan
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo, 693-8501, Japan
| | - Md Ahsanul Haque
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo, 693-8501, Japan
| | | | - Shuhei Yamaguchi
- Department of Internal Medicine III, Shimane University School of Medicine, 89-1 Enya Cho, Izumo, 693-8501, Japan
| |
Collapse
|
17
|
D'Amico RS, Zanazzi G, Wu P, Canoll P, Bruce JN. Pineal region glioblastomas display features of diffuse midline and non-midline gliomas. J Neurooncol 2018; 140:63-73. [PMID: 29992434 DOI: 10.1007/s11060-018-2931-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/17/2018] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Pineal region glioblastomas (GBM) are very rare, with approximately 46 cases described in the literature. The epidemiology, pathogenesis, and treatment of these lesions are poorly characterized. METHODS We identified all cases of pineal region GBM treated surgically at our institution between 1990 and 2017. Demographic and clinical follow-up data were extracted from the medical records for all cases. Pathology was reviewed and classified according to 2016 World Health Organization (WHO) criteria. Specific attention was given to the frequency of histone H3 K27M mutations in these midline gliomas. RESULTS Eight patients (seven men, one woman) with pineal region GBM, WHO grade IV, were identified. The most common presenting symptoms were headache (75%), vision changes (75%), and gait imbalance/ataxia (50%). Median age at diagnosis was 48.5 years (range 36-74 years). Radical subtotal resection, via a supracerebellar infratentorial approach, was achieved in 75% of patients. Review of the surgical pathology revealed seven primary GBMs (including one giant cell GBM) and one pineal region GBM that developed three years after resection of a pineal parenchymal tumor of intermediate differentiation. No cases demonstrated evidence of IDH-1 R132H mutation (N = 6) or 1p/19q co-deletion (N = 3). One case tested positive for the histone H3 K27M-mutation. Targeted exome sequencing of 467 cancer-related genes revealed nonsense mutations in ATRX and NF1. Adjuvant radiation and chemotherapy was employed in 87.5% and 75.0% of patients, respectively. Median overall survival (OS) was 15 months (range 2-24 months) from GBM diagnosis. CONCLUSIONS This study expands the clinical and pathologic spectrum of pineal region GBM, and provides the first report of the genetic landscape of these tumors.
Collapse
Affiliation(s)
- Randy S D'Amico
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA.
| | - George Zanazzi
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Peter Wu
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
18
|
Adams KV, Morshead CM. Neural stem cell heterogeneity in the mammalian forebrain. Prog Neurobiol 2018; 170:2-36. [PMID: 29902499 DOI: 10.1016/j.pneurobio.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 05/23/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022]
Abstract
The brain was long considered an organ that underwent very little change after development. It is now well established that the mammalian central nervous system contains neural stem cells that generate progeny that are capable of making new neurons, astrocytes, and oligodendrocytes throughout life. The field has advanced rapidly as it strives to understand the basic biology of these precursor cells, and explore their potential to promote brain repair. The purpose of this review is to present current knowledge about the diversity of neural stem cells in vitro and in vivo, and highlight distinctions between neural stem cell populations, throughout development, and within the niche. A comprehensive understanding of neural stem cell heterogeneity will provide insights into the cellular and molecular regulation of neural development and lifelong neurogenesis, and will guide the development of novel strategies to promote regeneration and neural repair.
Collapse
Affiliation(s)
- Kelsey V Adams
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada.
| | - Cindi M Morshead
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada; Department of Surgery, Division of Anatomy, Canada; Institute of Biomaterials and Biomedical Engineering, Canada; Rehabilitation Science Institute, University of Toronto, Canada.
| |
Collapse
|
19
|
Loubinoux I, Brihmat N, Castel-Lacanal E, Marque P. Cerebral imaging of post-stroke plasticity and tissue repair. Rev Neurol (Paris) 2017; 173:577-583. [DOI: 10.1016/j.neurol.2017.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/17/2023]
|
20
|
Preferential activation of HIF-2α adaptive signalling in neuronal-like cells in response to acute hypoxia. PLoS One 2017; 12:e0185664. [PMID: 28968430 PMCID: PMC5624621 DOI: 10.1371/journal.pone.0185664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/16/2017] [Indexed: 12/17/2022] Open
Abstract
Stroke causes severe neuronal damage as disrupted cerebral blood flow starves neurons of oxygen and glucose. The hypoxia inducible factors (HIF-1α and HIF-2α) orchestrate oxygen homeostasis and regulate specific aspects of hypoxic adaptation. Here we show the importance of HIF-2α dependant signalling in neuronal adaptation to hypoxic insult. PC12 and NT2 cells were differentiated into neuronal-like cells using NGF and retinoic acid, and exposed to acute hypoxia (1% O2). Gene and protein expression was analysed by qPCR and immunoblotting and the neuronal-like phenotype was examined. PC12 and NT2 differentiation promoted neurite extension and expression of neuronal markers, NSE and KCC2. Induction of HIF-1α mRNA or protein was not detected in hypoxic neuronal-like cells, however marked induction of HIF-2α mRNA and protein expression was observed. Induction of HIF-1α target genes was also not detected in response to acute hypoxia, however significant induction of HIF-2α transcriptional targets was clearly evident. Furthermore, hypoxic insult dramatically reduced both neurite number and length, and attenuated expression of neuronal markers, NSE and KCC2. This correlated with an increase in expression of the neural progenitor and stem cell-like markers, CD44 and vimentin, suggesting HIF-2α molecular mechanisms could potentially promote regression of neuronal-like cells to a stem-like state and trigger neuronal recovery following ischaemic insult. Our findings suggest the HIF-2α pathway predominates over HIF-1α signalling in neuronal-like cells following acute hypoxia.
Collapse
|
21
|
Abstract
Stroke is the leading cause of complex adult disability in the world. Recovery from stroke is often incomplete, which leaves many people dependent on others for their care. The improvement of long-term outcomes should, therefore, be a clinical and research priority. As a result of advances in our understanding of the biological mechanisms involved in recovery and repair after stroke, therapeutic opportunities to promote recovery through manipulation of poststroke plasticity have never been greater. This work has almost exclusively been carried out in preclinical animal models of stroke with little translation into human studies. The challenge ahead is to develop a mechanistic understanding of recovery from stroke in humans. Advances in neuroimaging techniques now enable us to reconcile behavioural accounts of recovery with molecular and cellular changes. Consequently, clinical trials can be designed in a stratified manner that takes into account when an intervention should be delivered and who is most likely to benefit. This approach is expected to lead to a substantial change in how restorative therapeutic strategies are delivered in patients after stroke.
Collapse
|
22
|
Senzacqua M, Severi I, Perugini J, Acciarini S, Cinti S, Giordano A. Action of Administered Ciliary Neurotrophic Factor on the Mouse Dorsal Vagal Complex. Front Neurosci 2016; 10:289. [PMID: 27445662 PMCID: PMC4921504 DOI: 10.3389/fnins.2016.00289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) induces weight loss in obese rodents and humans through activation of the hypothalamic Jak-STAT (Janus kinase-signal transducer and activator of transcription) signaling pathway. Here, we tested the hypothesis that CNTF also affects the brainstem centers involved in feeding and energy balance regulation. To this end, wild-type and leptin-deficient (ob/ob and db/db) obese mice were acutely treated with intraperitoneal recombinant CNTF. Coronal brainstem sections were processed for immunohistochemical detection of STAT3, STAT1, STAT5 phosphorylation and c-Fos. In wild-type mice, CNTF treatment for 45 min induced STAT3, STAT1, and STAT5 phosphorylation in neurons as well as glial cells of the area postrema; here, the majority of CNTF-responsive cells activated multiple STAT isoforms, and a significant proportion of CNTF-responsive glial cells bore the immaturity and plasticity markers nestin and vimentin. After 120 min CNTF treatment, c-Fos expression was intense in glial cells and weak in neurons of the area postrema, it was intense in several neurons of the rostral and caudal solitary tract nucleus (NTS), and weak in some cholinergic neurons of the dorsal motor nucleus of the vagus. In the ob/ob and db/db mice, Jak-STAT activation and c-Fos expression were similar to those induced in wild-type mouse brainstem. Treatment with CNTF (120 min, to induce c-Fos expression) and leptin (25 min, to induce STAT3 phosphorylation) demonstrated the co-localization of the two transcription factors in a small neuron population in the caudal NTS portion. Finally, weak immunohistochemical CNTF staining, detected in funiculus separans, and meningeal glial cells, matched the modest amount of CNTF found by RT-qPCR in micropunched area postrema tissue, which in contrast exhibited a very high amount of CNTF receptor. Collectively, the present findings show that the area postrema and the NTS exhibit high, distinctive responsiveness to circulating exogenous and, probably, endogenous CNTF.
Collapse
Affiliation(s)
- Martina Senzacqua
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche Ancona, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche Ancona, Italy
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche Ancona, Italy
| | - Samantha Acciarini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy; Center of Obesity, Università Politecnica delle Marche-United HospitalsAncona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche Ancona, Italy
| |
Collapse
|
23
|
Carmichael ST. Emergent properties of neural repair: elemental biology to therapeutic concepts. Ann Neurol 2016; 79:895-906. [PMID: 27043816 PMCID: PMC4884133 DOI: 10.1002/ana.24653] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 03/28/2016] [Accepted: 03/29/2016] [Indexed: 12/20/2022]
Abstract
Stroke is the leading cause of adult disability. The past decade has seen advances in basic science research of neural repair in stroke. The brain forms new connections after stroke, which have a causal role in recovery of function. Brain progenitors, including neuronal and glial progenitors, respond to stroke and initiate a partial formation of new neurons and glial cells. The molecular systems that underlie axonal sprouting, neurogenesis, and gliogenesis after stroke have recently been identified. Importantly, tractable drug targets exist within these molecular systems that might stimulate tissue repair. These basic science advances have taken the field to its first scientific milestone; the elemental principles of neural repair in stroke have been identified. The next stages in this field involve understanding how these elemental principles of recovery interact in the dynamic cellular systems of the repairing brain. Emergent principles arise out of the interaction of the fundamental or elemental principles in a system. In neural repair, the elemental principles of brain reorganization after stroke interact to generate higher order and distinct concepts of regenerative brain niches in cellular repair, neuronal networks in synaptic plasticity, and the distinction of molecular systems of neuroregeneration. Many of these emergent principles directly guide the development of new therapies, such as the necessity for spatial and temporal control in neural repair therapy delivery and the overlap of cancer and neural repair mechanisms. This review discusses the emergent principles of neural repair in stroke as they relate to scientific and therapeutic concepts in this field. Ann Neurol 2016;79:895–906
Collapse
Affiliation(s)
- S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA and UCLA Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
24
|
Marlier Q, Verteneuil S, Vandenbosch R, Malgrange B. Mechanisms and Functional Significance of Stroke-Induced Neurogenesis. Front Neurosci 2015; 9:458. [PMID: 26696816 PMCID: PMC4672088 DOI: 10.3389/fnins.2015.00458] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023] Open
Abstract
Stroke affects one in every six people worldwide, and is the leading cause of adult disability. After stroke, some limited spontaneous recovery occurs, the mechanisms of which remain largely unknown. Multiple, parallel approaches are being investigated to develop neuroprotective, reparative and regenerative strategies for the treatment of stroke. For years, clinical studies have tried to use exogenous cell therapy as a means of brain repair, with varying success. Since the rediscovery of adult neurogenesis and the identification of adult neural stem cells in the late nineties, one promising field of investigation is focused upon triggering and stimulating this self-repair system to replace the neurons lost following brain injury. For instance, it is has been demonstrated that the adult brain has the capacity to produce large numbers of new neurons in response to stroke. The purpose of this review is to provide an updated overview of stroke-induced adult neurogenesis, from a cellular and molecular perspective, to its impact on brain repair and functional recovery.
Collapse
Affiliation(s)
- Quentin Marlier
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman Liege, Belgium
| | | | - Renaud Vandenbosch
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman Liege, Belgium
| | - Brigitte Malgrange
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman Liege, Belgium
| |
Collapse
|
25
|
Lin R, Iacovitti L. Classic and novel stem cell niches in brain homeostasis and repair. Brain Res 2015; 1628:327-342. [DOI: 10.1016/j.brainres.2015.04.029] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023]
|
26
|
Miyata S. New aspects in fenestrated capillary and tissue dynamics in the sensory circumventricular organs of adult brains. Front Neurosci 2015; 9:390. [PMID: 26578857 PMCID: PMC4621430 DOI: 10.3389/fnins.2015.00390] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/05/2015] [Indexed: 12/20/2022] Open
Abstract
The blood-brain barrier (BBB) generally consists of endothelial tight junction barriers that prevent the free entry of blood-derived substances, thereby maintaining the extracellular environment of the brain. However, the circumventricular organs (CVOs), which are located along the midlines of the brain ventricles, lack these endothelial barriers and have fenestrated capillaries; therefore, they have a number of essential functions, including the transduction of information between the blood circulation and brain. Previous studies have demonstrated the extensive contribution of the CVOs to body fluid and thermal homeostasis, energy balance, the chemoreception of blood-derived substances, and neuroinflammation. In this review, recent advances have been discussed in fenestrated capillary characterization and dynamic tissue reconstruction accompanied by angiogenesis and neurogliogenesis in the sensory CVOs of adult brains. The sensory CVOs, including the organum vasculosum of the lamina terminalis (OVLT), subfornical organ (SFO), and area postrema (AP), have size-selective and heterogeneous vascular permeabilities. Astrocyte-/tanycyte-like neural stem cells (NSCs) sense blood- and cerebrospinal fluid-derived information through the transient receptor potential vanilloid 1, a mechanical/osmotic receptor, Toll-like receptor 4, a lipopolysaccharide receptor, and Nax, a Na-sensing Na channel. They also express tight junction proteins and densely and tightly surround mature neurons to protect them from blood-derived neurotoxic substances, indicating that the NSCs of the CVOs perform BBB functions while maintaining the capacity to differentiate into new neurons and glial cells. In addition to neurogliogenesis, the density of fenestrated capillaries is regulated by angiogenesis, which is accompanied by the active proliferation and sprouting of endothelial cells. Vascular endothelial growth factor (VEGF) signaling may be involved in angiogenesis and neurogliogenesis, both of which affect vascular permeability. Thus, recent findings advocate novel concepts for the CVOs, which have the dynamic features of vascular and parenchymal tissues.
Collapse
Affiliation(s)
- Seiji Miyata
- Department of Applied Biology, Kyoto Institute of TechnologyKyoto, Japan
| |
Collapse
|
27
|
Konieczny MJ, Ri SJ, Georgiadis JR. Omental Approach to Functional Recovery After Cerebrovascular Disease. World Neurosurg 2015; 87:406-16. [PMID: 26493716 DOI: 10.1016/j.wneu.2015.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/05/2015] [Accepted: 10/07/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To review and synthesize the clinical literature regarding risks and benefits of omentum transplantation and transposition surgery in patients with ischemic stroke of other etiology (non-MMD) and Moyamoya disease (MMD), and to evaluate the evidence for biological underpinnings of the presumed physiologic effects of omentum transplantation and transposition on vascularization of brain parenchyma. METHODS Articles were searched on scientific databases using predefined key terms. Data abstraction was based on the clinical course as reported in the articles. For further analysis, patients were divided into groups according to their diagnosis (MMD or non-MMD). Descriptive statistics were computed for better integration of the results. RESULTS The final literature review contained 15 articles (11 case series, 4 single case studies) with data on 93 patients (29 non-MMD, 64 MMD). At post-assessment 56% of patients showed substantial gains in functional domains (24% in the non-MMD group, 71% in the MMD group) and 92% demonstrated improvements of cerebral vascularization (55% in the non-MMD group, 98% in the MMD group). Differences in improvement became apparent with regard to the initial symptomatology wherein transient ischemic attacks were related to superior recovery rates and language pathologies showed least improvement. CONCLUSIONS Surgical revascularization using omental tissue has shown good success rates, particularly for recurrent transient ischemic attacks and prevention of further strokes and should be considered as treatment option for selected patients. Experimental data on the physiologic basis for postoperative improvement delivered convincing evidence for its arteriogenic potential and recent developments in omental stem cell research suggest a role in recovery from long-standing neurological deficits.
Collapse
Affiliation(s)
- Marek J Konieczny
- Department of Molecular Neurobiology, Graduate School of Behavioural and Cognitive Neurosciences, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Charite-Universitätsmedizin Berlin, Department of Experimental Neurology, Campus Benjamin Franklin, Berlin, Germany.
| | - Song-Jin Ri
- Charite-Universitätsmedizin Berlin, Department of Experimental Neurology, Campus Benjamin Franklin, Berlin, Germany
| | - Janniko R Georgiadis
- Department of Neuroscience, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
28
|
Furube E, Morita M, Miyata S. Characterization of neural stem cells and their progeny in the sensory circumventricular organs of adult mouse. Cell Tissue Res 2015; 362:347-65. [PMID: 25994374 DOI: 10.1007/s00441-015-2201-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/11/2015] [Indexed: 01/19/2023]
Abstract
Although evidence has accumulated that neurogenesis and gliogenesis occur in the subventricular zone (SVZ) and subgranular zone (SGZ) of adult mammalian brains, recent studies indicate the presence of neural stem cells (NSCs) in adult brains, particularly the circumventricular regions. In the present study, we aimed to determine characterization of NSCs and their progenitor cells in the sensory circumventricular organs (CVOs), including organum vasculosum of the lamina terminalis, subfornical organ, and area postrema of adult mouse. There were two types of NSCs: tanycyte-like ependymal cells and astrocyte-like cells. Astrocyte-like NSCs proliferated slowly and oligodendrocyte progenitor cells (OPCs) and neural progenitor cells (NPCs) actively divided. Molecular marker protein expression of NSCs and their progenitor cells were similar to those reported in the SVZ and SGZ, except that astrocyte-like NSCs expressed S100β. These circumventricular NSCs possessed the capacity to give rise to oligodendrocytes and sparse numbers of neurons and astrocytes in the sensory CVOs and adjacent brain regions. The inhibition of vascular endothelial growth factor (VEGF) signaling by using a VEGF receptor-associated tyrosine kinase inhibitor AZD2171 largely suppressed basal proliferation of OPCs. A single systemic administration of lipopolysaccharide attenuated proliferation of OPCs and induced remarkable proliferation of microglia. The present study indicates that sensory circumventricular NSCs provide new neurons and glial cells in the sensory CVOs and adjacent brain regions.
Collapse
Affiliation(s)
- Eriko Furube
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, 606-8585, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, 606-8585, Japan.
| |
Collapse
|
29
|
Pócsai K, Kálmán M. Glial and perivascular structures in the subfornical organ: distinguishing the shell and core. J Histochem Cytochem 2015; 63:367-83. [PMID: 25673286 DOI: 10.1369/0022155415575027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 01/25/2015] [Indexed: 11/22/2022] Open
Abstract
The subfornical organ (SFO) is a circumventricular organ with a chemosensitive function, and its vessels have no blood-brain barrier. Our study investigated the glial and vascular components in the SFO to determine whether their distributions indicate subdivisions, how to characterize the vessels and how to demarcate the SFO. To this end, we investigated glial markers (GFAP, glutamine synthetase, S100) and other markers, including vimentin and nestin (immature glia), laminin (basal lamina), β-dystroglycan (glio-vascular connections), and aquaporin 4 (glial water channels). We determined that the 'shell' of the SFO was marked by immunoreactivity for S100, GFAP and aquaporin 4. Nestin immunoreactivity was characteristic of the 'core'. Vimentin was almost evenly distributed. Glutamine synthetase immunoreactivity occurred in the shell but its expression was sparse. Vessels in the core were decorated with laminin but showed a discontinuous expression of aquaporin 4. Vimentin and GFAP staining was usually in separate glial elements, which may be related to their functional differences. Similar to other vessels in the brain, β-dystroglycan was detected along the shell vessels but laminin was not. The gradual disappearance of the laminin immunopositivity was attributed to the gradual disappearance of the perivascular space. Thus, our findings suggest that the shell and core glio-vascular structures are adapted to different sensory functions: osmoperception and the perception of circulating peptides, respectively.
Collapse
Affiliation(s)
- Károly Pócsai
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary (KP, MK)
| | - Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary (KP, MK)
| |
Collapse
|
30
|
Lin R, Cai J, Nathan C, Wei X, Schleidt S, Rosenwasser R, Iacovitti L. Neurogenesis is enhanced by stroke in multiple new stem cell niches along the ventricular system at sites of high BBB permeability. Neurobiol Dis 2015; 74:229-39. [DOI: 10.1016/j.nbd.2014.11.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/03/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023] Open
|
31
|
Kleine TO. Cellular immune surveillance of central nervous system bypasses blood-brain barrier and blood-cerebrospinal-fluid barrier: Revealed with the New Marburg cerebrospinal-fluid model in healthy humans. Cytometry A 2015; 87:227-43. [DOI: 10.1002/cyto.a.22589] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/21/2014] [Indexed: 01/12/2023]
Affiliation(s)
- Tilmann O. Kleine
- Department of Laboratory Medicine and Molecular Diagnostics of the University Hospital Marburg. Dependance: Cerebrospinal-Fluid References Labor, Baldingerstraße; 35043 Marburg Germany
| |
Collapse
|
32
|
Yang X, Zhu F, Zhang X, Gao Z, Cao Y. Ipsilateral versus bilateral limb-training in promoting the proliferation and differentiation of endogenous neural stem cells following cerebral infarction in rats. Neural Regen Res 2014; 7:2698-704. [PMID: 25337116 PMCID: PMC4200738 DOI: 10.3969/j.issn.1673-5374.2012.34.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 11/02/2012] [Indexed: 12/11/2022] Open
Abstract
We investigated the effects of ipsilateral versus bilateral limb-training on promotion of endogenous neural stem cells in the peripheral infarct zone and the corresponding cerebral region in the unaffected hemisphere of rats with cerebral infarction. Middle cerebral artery occlusion was induced in Wistar rats. The rat forelimb on the unaffected side was either wrapped up with tape to force the use of the paretic forelimb in rats or not braked to allow bilateral forelimbs to participate in training. Daily training consisted of mesh drum training, balance beam training, and stick rolling training for a total of 40 minutes, once per day. Control rats received no training. At 14 days after functional training, rats receiving bilateral limb-training exhibited milder neurological impairment than that in the ipsilateral limb-training group or the control group. The number of nestin/glial fibrillary acidic protein-positive and nestin/microtubule-associated protein 2-positive cells in the peripheral infarct zone and in the corresponding cerebral region in the unaffected hemisphere was significantly higher in rats receiving bilateral limb-training than in rats receiving ipsilateral limb-training. These data suggest that bilateral limb-training can promote the proliferation and differentiation of endogenous neural stem cells in the bilateral hemispheres after cerebral infarction and accelerate the recovery of neurologic function. In addition, bilateral limb-training produces better therapeutic effects than ipsilateral limb-training.
Collapse
Affiliation(s)
- Xiyao Yang
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Feng Zhu
- Department of Neurology, Shenyang Brain Hospital, Shenyang 110001, Liaoning Province, China
| | - Xiaomei Zhang
- The First People's Hospital of Dandong, Dandong 118000, Liaoning Province, China
| | - Zhuo Gao
- Department of Neurology, Shenyang Brain Hospital, Shenyang 110001, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
33
|
Edebali N, Tekin IÖ, Açıkgöz B, Açıkgöz S, Barut F, Sevinç N, Sümbüloğlu V. Apoptosis and necrosis in the circumventricular organs after experimental subarachnoid hemorrhage as detected with annexin V and caspase 3 immunostaining. Neurol Res 2014; 36:1114-20. [PMID: 25137492 DOI: 10.1179/1743132814y.0000000437] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES The circumventricular organs (CVOs) are essential for most autonomic and endocrine functions. Trauma and bleeding can affect their function. The aim of this study was to investigate apoptosis and necrosis in CVOs in the early period after experimental subarachnoid hemorrhage (SAH) in rats, using annexin V affinity and caspase 3 immunostaining. METHODS Three experimental groups were used: Days 1 and 2 after SAH, and a control group, seven Wistar albino rats each. Subarachnoid hemorrhage was accomplished by transclival basilar artery puncture. Rats were perfused with 0.9% NaCl and 0·1M phosphate buffer pH 7.4 until heart stoppage. Apoptosis and necrosis in CVOs were measured by flow cytometry with annexin V staining, and by caspase 3 immunostaining. RESULTS Apoptosis in the organum vasculosum lamina terminalis (OVLT), median eminence (ME), and area postrema (AP) was significantly higher in the Day 1 group than in the control group. Apoptosis in the subfornicial organ (SFO), OVLT, ME, and AP was significantly higher in the Day 2 group than in the control group. There were significant differences between the Day 1 and Day 2 groups, except for AP. Necrosis in SFO and OVLT was significantly higher in the Day 2 group than in the Day 1 or control groups, whereas necrosis in the ME and AP did not differ between the three groups. Caspase 3-positive cell density was more intense in the Day 2 group than in the Day 1 and control groups. DISCUSSION Prevention of apoptosis may potentially improve impaired functions of CVOs after SAH.
Collapse
|
34
|
Teissier N, Fallet-Bianco C, Delezoide AL, Laquerrière A, Marcorelles P, Khung-Savatovsky S, Nardelli J, Cipriani S, Csaba Z, Picone O, Golden JA, Van Den Abbeele T, Gressens P, Adle-Biassette H. Cytomegalovirus-induced brain malformations in fetuses. J Neuropathol Exp Neurol 2014; 73:143-58. [PMID: 24423639 DOI: 10.1097/nen.0000000000000038] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Neurologic morbidity associated with congenital cytomegalovirus (CMV) infection is a major public health concern. The pathogenesis of cerebral lesions remains unclear. We report the neuropathologic substrates, the immune response, and the cellular targets of CMV in 16 infected human fetal brains aged 23 to 28.5 gestational weeks. Nine cases were microcephalic, 10 had extensive cortical lesions, 8 had hippocampal abnormalities, and 5 cases showed infection of the olfactory bulb. The density of CMV-immunolabeled cells correlated with the presence of microcephaly and the extent of brain abnormalities. Innate and adaptive immune responses were present but did not react against all CMV-infected cells. Cytomegalovirus infected all cell types but showed higher tropism for stem cells/radial glial cells. The results indicate that 2 main factors influence the neuropathologic outcome at this stage: the density of CMV-positive cells and the tropism of CMV for stem/progenitor cells. This suggests that the large spectrum of CMV-induced brain abnormalities is caused not only by tissue destruction but also by the particular vulnerability of stem cells during early brain development. Florid infection of the hippocampus and the olfactory bulb may expose these patients to the risk of neurocognitive and sensorineural handicap even in cases of infection at late stages of gestation.
Collapse
Affiliation(s)
- Natacha Teissier
- From the Inserm (NT, A-LD, SK-S, JN, SC, ZC, TVDA, PG, HA-B); Univ Paris Diderot, Sorbonne Paris Cité (NT, A-LD, S-KS, JN, SC, TVDA, PG, HAB); Paediatric Otorhinolaryngology Department, Robert Debré Hospital (NT, TVDA); Department of Pathology, Sainte-Anne/Cochin Hospital (CF-B, A-LD); and Biology of Development Department, Robert Debré Hospital (A-LD, SK-S), Paris; Department of Pathology, Charles Nicolle Hospital, Rouen (AL); Department of Pathology, Morvan Hospital, Brest (PM); Obstetrics Department, Béclère Hospital, Clamart (OP); and Obstetrics Department, Foch Hospital, Suresnes (OP), France; Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (JAG); Centre for the Developing Brain, King's College, St. Thomas' Campus, London, United Kingdom (PG); and Department of Pathology, Lariboisière Hospital, Paris, France (HA-B)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|