51
|
Hypothalamic Astrocytes as a Specialized and Responsive Cell Population in Obesity. Int J Mol Sci 2021; 22:ijms22126176. [PMID: 34201099 PMCID: PMC8228119 DOI: 10.3390/ijms22126176] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are a type of glial cell anatomically and functionally integrated into the neuronal regulatory circuits for the neuroendocrine control of metabolism. Being functional integral compounds of synapses, astrocytes are actively involved in the physiological regulatory aspects of metabolic control, but also in the pathological processes that link neuronal dysfunction and obesity. Between brain areas, the hypothalamus harbors specialized functional circuits that seem selectively vulnerable to metabolic damage, undergoing early cellular rearrangements which are thought to be at the core of the pathogenesis of diet-induced obesity. Such changes in the hypothalamic brain region consist of a rise in proinflammatory cytokines, the presence of a reactive phenotype in astrocytes and microglia, alterations in the cytoarchitecture and synaptology of hypothalamic circuits, and angiogenesis, a phenomenon that cannot be found elsewhere in the brain. Increasing evidence points to the direct involvement of hypothalamic astrocytes in such early metabolic disturbances, thus moving the study of these glial cells to the forefront of obesity research. Here we provide a comprehensive review of the most relevant findings of molecular and pathophysiological mechanisms by which hypothalamic astrocytes might be involved in the pathogenesis of obesity.
Collapse
|
52
|
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: 0.5] [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
|
53
|
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
|
54
|
Dardente H, Migaud M. Thyroid hormone and hypothalamic stem cells in seasonal functions. VITAMINS AND HORMONES 2021; 116:91-131. [PMID: 33752829 DOI: 10.1016/bs.vh.2021.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Seasonal rhythms are a pervasive feature of most living organisms, which underlie yearly timeliness in breeding, migration, hibernation or weight gain and loss. To achieve this, organisms have developed inner timing devices (circannual clocks) that endow them with the ability to predict then anticipate changes to come, usually using daylength as the proximate cue. In Vertebrates, daylength interpretation involves photoperiodic control of TSH production by the pars tuberalis (PT) of the pituitary, which governs a seasonal switch in thyroid hormone (TH) availability in the neighboring hypothalamus. Tanycytes, specialized glial cells lining the third ventricle (3V), are responsible for this TH output through the opposite, PT-TSH-driven, seasonal control of deiodinases 2/3 (Dio 2/3). Tanycytes comprise a photoperiod-sensitive stem cell niche and TH is known to play major roles in cell proliferation and differentiation, which suggests that seasonal control of tanycyte proliferation may be involved in the photoperiodic synchronization of seasonal rhythms. Here we review our current knowledge of the molecular and neuroendocrine pathway linking photoperiodic information to seasonal changes in physiological functions and discuss the potential implication of tanycytes, TH and cell proliferation in seasonal timing.
Collapse
Affiliation(s)
- Hugues Dardente
- PRC, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France.
| | - Martine Migaud
- PRC, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
55
|
Kaya M, Ahishali B. Basic physiology of the blood-brain barrier in health and disease: a brief overview. Tissue Barriers 2021; 9:1840913. [PMID: 33190576 PMCID: PMC7849738 DOI: 10.1080/21688370.2020.1840913] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022] Open
Abstract
The blood-brain barrier (BBB), a dynamic interface between blood and brain constituted mainly by endothelial cells of brain microvessels, robustly restricts the entry of potentially harmful blood-sourced substances and cells into the brain, however, many therapeutically active agents concurrently cannot gain access into the brain at effective doses in the presence of an intact barrier. On the other hand, breakdown of BBB integrity may involve in the pathogenesis of various neurodegenerative diseases. Besides, certain diseases/disorders such as Alzheimer's disease, hypertension, and epilepsy are associated with varying degrees of BBB disruption. In this review, we aim to highlight the current knowledge on the cellular and molecular composition of the BBB with special emphasis on the major transport pathways across the barrier type endothelial cells. We further provide a discussion on the innovative brain drug delivery strategies in which the obstacle formed by BBB interferes with effective pharmacological treatment of neurodegenerative diseases/disorders.
Collapse
Affiliation(s)
- Mehmet Kaya
- Koç University School of Medicine Department of Physiology, Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Bulent Ahishali
- Koç University School of Medicine Department of Histology and Embryology, Koç University Research Center for Translational Medicine, Istanbul, Turkey
| |
Collapse
|
56
|
Ragozzino FJ, Arnold RA, Kowalski CW, Savenkova MI, Karatsoreos IN, Peters JH. Corticosterone inhibits vagal afferent glutamate release in the nucleus of the solitary tract via retrograde endocannabinoid signaling. Am J Physiol Cell Physiol 2020; 319:C1097-C1106. [PMID: 32966126 DOI: 10.1152/ajpcell.00190.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Circulating blood glucocorticoid levels are dynamic and responsive to stimuli that impact autonomic function. In the brain stem, vagal afferent terminals release the excitatory neurotransmitter glutamate to neurons in the nucleus of the solitary tract (NTS). Vagal afferents integrate direct visceral signals and circulating hormones with ongoing NTS activity to control autonomic function and behavior. Here, we investigated the effects of corticosterone (CORT) on glutamate signaling in the NTS using patch-clamp electrophysiology on brain stem slices containing the NTS and central afferent terminals from male C57BL/6 mice. We found that CORT rapidly decreased both action potential-evoked and spontaneous glutamate signaling. The effects of CORT were phenocopied by dexamethasone and blocked by mifepristone, consistent with glucocorticoid receptor (GR)-mediated signaling. While mRNA for GR was present in both the NTS and vagal afferent neurons, selective intracellular quenching of G protein signaling in postsynaptic NTS neurons eliminated the effects of CORT. We then investigated the contribution of retrograde endocannabinoid signaling, which has been reported to transduce nongenomic GR effects. Pharmacological or genetic elimination of the cannabinoid type 1 receptor signaling blocked CORT suppression of glutamate release. Together, our results detail a mechanism, whereby the NTS integrates endocrine CORT signals with fast neurotransmission to control autonomic reflex pathways.
Collapse
Affiliation(s)
- Forrest J Ragozzino
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Rachel A Arnold
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Cody W Kowalski
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Marina I Savenkova
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington.,Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - James H Peters
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
57
|
Fenrich M, Habjanovic K, Kajan J, Heffer M. The circle of Willis revisited: Forebrain dehydration sensing facilitated by the anterior communicating artery: How hemodynamic properties facilitate more efficient dehydration sensing in amniotes. Bioessays 2020; 43:e2000115. [PMID: 33191609 DOI: 10.1002/bies.202000115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022]
Abstract
We hypothesize that threat of dehydration provided selection pressure for the evolutionary emergence and persistence of the anterior communicating artery (ACoA - the inter-arterial connection that completes the Circle of Willis) in early amniotes. The ACoA is a hemodynamically insignificant artery, but, as we argue in this paper, its privileged position outside the blood-brain barrier gives it a crucial sensing function for the osmolarity of the blood against the background of the rest of the brain, which efficiently protects itself from dehydration. Till now, the questions of why the ACoA evolved, and what its physiological function is, have remained unsatisfactorily answered. The traditional view-that the ACoA serves as a collateral source of vascularization in case of arterial stenosis-is anthropocentric, and not in accordance with principles of natural selection that apply more generally. Diseases underlying arterial stenosis are associated with aging and the human lifestyle, so this cannot explain why the ACoA formed hundreds of millions of years ago and persisted in amniotes to this day. The peculiar hemodynamic properties of the ACoA could be selected traits that allowed for more efficient forebrain detection of dehydration and complex behavioral responses to water loss, a major advantage in the survival of early amniotes. This hypothesis also explains insufficient hydration often seen in elderly humans.
Collapse
Affiliation(s)
- Matija Fenrich
- Laboratory of Neurobiology, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Karlo Habjanovic
- Laboratory of Neurobiology, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Josip Kajan
- Laboratory of Neurobiology, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Marija Heffer
- Laboratory of Neurobiology, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
58
|
Profaci CP, Munji RN, Pulido RS, Daneman R. The blood-brain barrier in health and disease: Important unanswered questions. J Exp Med 2020; 217:151582. [PMID: 32211826 PMCID: PMC7144528 DOI: 10.1084/jem.20190062] [Citation(s) in RCA: 422] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/21/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
The blood vessels vascularizing the central nervous system exhibit a series of distinct properties that tightly control the movement of ions, molecules, and cells between the blood and the parenchyma. This "blood-brain barrier" is initiated during angiogenesis via signals from the surrounding neural environment, and its integrity remains vital for homeostasis and neural protection throughout life. Blood-brain barrier dysfunction contributes to pathology in a range of neurological conditions including multiple sclerosis, stroke, and epilepsy, and has also been implicated in neurodegenerative diseases such as Alzheimer's disease. This review will discuss current knowledge and key unanswered questions regarding the blood-brain barrier in health and disease.
Collapse
Affiliation(s)
- Caterina P Profaci
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Roeben N Munji
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Robert S Pulido
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Richard Daneman
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| |
Collapse
|
59
|
Verheggen ICM, de Jong JJA, van Boxtel MPJ, Postma AA, Verhey FRJ, Jansen JFA, Backes WH. Permeability of the windows of the brain: feasibility of dynamic contrast-enhanced MRI of the circumventricular organs. Fluids Barriers CNS 2020; 17:66. [PMID: 33115484 PMCID: PMC7594295 DOI: 10.1186/s12987-020-00228-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Circumventricular organs (CVOs) are small structures without a blood-brain barrier surrounding the brain ventricles that serve homeostasic functions and facilitate communication between the blood, cerebrospinal fluid and brain. Secretory CVOs release peptides and sensory CVOs regulate signal transmission. However, pathogens may enter the brain through the CVOs and trigger neuroinflammation and neurodegeneration. We investigated the feasibility of dynamic contrast-enhanced (DCE) MRI to assess the CVO permeability characteristics in vivo, and expected significant contrast uptake in these regions, due to blood-brain barrier absence. METHODS Twenty healthy, middle-aged to older males underwent brain DCE MRI. Pharmacokinetic modeling was applied to contrast concentration time-courses of CVOs, and in reference to white and gray matter. We investigated whether a significant and positive transfer from blood to brain could be measured in the CVOs, and whether this differed between secretory and sensory CVOs or from normal-appearing brain matter. RESULTS In both the secretory and sensory CVOs, the transfer constants were significantly positive, and all secretory CVOs had significantly higher transfer than each sensory CVO. The transfer constants in both the secretory and sensory CVOs were higher than in the white and gray matter. CONCLUSIONS Current measurements confirm the often-held assumption of highly permeable CVOs, of which the secretory types have the strongest blood-to-brain transfer. The current study suggests that DCE MRI could be a promising technique to further assess the function of the CVOs and how pathogens can potentially enter the brain via these structures. TRIAL REGISTRATION Netherlands Trial Register number: NL6358, date of registration: 2017-03-24.
Collapse
Affiliation(s)
- Inge C M Verheggen
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
- Alzheimer Center Limburg, Maastricht, The Netherlands.
| | - Joost J A de Jong
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Alida A Postma
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
60
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
61
|
Non-Human Primate Blood-Brain Barrier and In Vitro Brain Endothelium: From Transcriptome to the Establishment of a New Model. Pharmaceutics 2020; 12:pharmaceutics12100967. [PMID: 33066641 PMCID: PMC7602447 DOI: 10.3390/pharmaceutics12100967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
The non-human primate (NHP)-brain endothelium constitutes an essential alternative to human in the prediction of molecule trafficking across the blood–brain barrier (BBB). This study presents a comparison between the NHP transcriptome of freshly isolated brain microcapillaries and in vitro-selected brain endothelial cells (BECs), focusing on important BBB features, namely tight junctions, receptors mediating transcytosis (RMT), ABC and SLC transporters, given its relevance as an alternative model for the molecule trafficking prediction across the BBB and identification of new brain-specific transport mechanisms. In vitro BECs conserved most of the BBB key elements for barrier integrity and control of molecular trafficking. The function of RMT via the transferrin receptor (TFRC) was characterized in this NHP-BBB model, where both human transferrin and anti-hTFRC antibody showed increased apical-to-basolateral passage in comparison to control molecules. In parallel, eventual BBB-related regional differences were Investig.igated in seven-day in vitro-selected BECs from five brain structures: brainstem, cerebellum, cortex, hippocampus, and striatum. Our analysis retrieved few differences in the brain endothelium across brain regions, suggesting a rather homogeneous BBB function across the brain parenchyma. The presently established NHP-derived BBB model closely mimics the physiological BBB, thus representing a ready-to-use tool for assessment of the penetration of biotherapeutics into the human CNS.
Collapse
|
62
|
Tauber SC, Djukic M, Gossner J, Eiffert H, Brück W, Nau R. Sepsis-associated encephalopathy and septic encephalitis: an update. Expert Rev Anti Infect Ther 2020; 19:215-231. [PMID: 32808580 DOI: 10.1080/14787210.2020.1812384] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Sepsis-associated encephalopathy (SAE) and septic encephalitis (SE) are associated with increased mortality, long-term cognitive impairment, and focal neurological deficits. AREAS COVERED The PUBMED database was searched 2016-2020. The clinical manifestation of SAE is delirium, SE additionally is characterized by focal neurological symptoms. SAE is caused by inflammation with endothelial/microglial activation, increase of permeability of the blood-brain-barrier, hypoxia, imbalance of neurotransmitters, glial activation, axonal, and neuronal loss. Septic-embolic (SEE) and septic-metastatic encephalitis (SME) are characterized by focal ischemia (SEE) and small abscesses (SME). The continuum between SAE, SME, and SEE is documented by imaging techniques and autopsies. The backbone of treatment is rapid optimum antibiotic therapy. Experimental approaches focus on modulation of inflammation, stabilization of the blood-brain barrier, and restoration of membrane/mitochondrial function. EXPERT OPINION The most promising diagnostic approaches are new imaging techniques. The most important measure to fight delirium remains establishment of daily structure and adequate sensory stimuli. Dexmedetomidine and melatonin appear to reduce the frequency of delirium, their efficacy in SAE and SE remains to be established. Drugs already licensed for other indications or available as food supplements which may be effective in SAE are statins, L-DOPA/benserazide, β-hydroxybutyrate, palmitoylethanolamide, and tetracyclines or other bactericidal non-lytic antibiotics.
Collapse
Affiliation(s)
- Simone C Tauber
- Department of Neurology, Rheinisch-Westfälische Technische Hochschule (RWTH) , Aachen, Germany
| | - Marija Djukic
- Institute of Neuropathology, University Medical Center , Göttingen, Germany.,Department of Geriatrics, Protestant Hospital Göttingen-Weende , Göttingen, Germany
| | - Johannes Gossner
- Department of Diagnostic and Interventional Radiology, Protestant Hospital Göttingen-Weende , Göttingen, Germany
| | - Helmut Eiffert
- Amedes MVZ for Laboratory Medicine, Medical Microbiology and Infectiology , Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center , Göttingen, Germany
| | - Roland Nau
- Institute of Neuropathology, University Medical Center , Göttingen, Germany.,Department of Geriatrics, Protestant Hospital Göttingen-Weende , Göttingen, Germany
| |
Collapse
|
63
|
Cobo-Calvo A, Ruiz A, Richard C, Blondel S, Cavagna S, Strazielle N, Ghersi-Egea JF, Giraudon P, Marignier R. Purified IgG from aquaporin-4 neuromyelitis optica spectrum disorder patients alters blood-brain barrier permeability. PLoS One 2020; 15:e0238301. [PMID: 32881954 PMCID: PMC7470361 DOI: 10.1371/journal.pone.0238301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Neuromyelitis optica spectrum disorders (NMOSD) is a primary astrocytopathy driven by antibodies directed against the aquaporin-4 water channel located at the end-feet of the astrocyte. Although blood-brain barrier (BBB) breakdown is considered one of the key steps for the development and lesion formation, little is known about the molecular mechanisms involved. The aim of the study was to evaluate the effect of human immunoglobulins from NMOSD patients (NMO-IgG) on BBB properties. METHODS Freshly isolated brain microvessels (IBMs) from rat brains were used as a study model. At first, analysis of the secretome profile from IBMs exposed to purified NMO-IgG, to healthy donor IgG (Control-IgG), or non-treated, was performed. Second, tight junction (TJ) proteins expression in fresh IBMs and primary cultures of brain microvascular endothelial cells (BMEC) was analysed by Western blotting (Wb) after exposition to NMO-IgG and Control-IgG. Finally, functional BBB properties were investigated evaluating the presence of rat-IgG in tissue lysate from brain using Wb in the rat-model, and the passage of NMO-IgG and sucrose in a bicameral model. RESULTS We found that NMO-IgG induces functional and morphological BBB changes, including: 1) increase of pro-inflammatory cytokines production (CXCL-10 [IP-10], IL-6, IL-1RA, IL-1β and CXCL-3) in IBMs when exposed to NMO-IgG; 2) decrease of Claudin-5 levels by 25.6% after treatment of fresh IBMs by NMO-IgG compared to Control-IgG (p = 0.002), and similarly, decrease of Claudin-5 by at least 20% when BMEC were cultured with NMO-IgG from five different patients; 3) a higher level of rat-IgG accumulated in periventricular regions of NMO-rats compared to Control-rats and an increase in the permeability of BBB after NMO-IgG treatment in the bicameral model. CONCLUSION Human NMO-IgG induces both structural and functional alterations of BBB properties, suggesting a direct role of NMO-IgG on modulation of BBB permeability in NMOSD.
Collapse
Affiliation(s)
- Alvaro Cobo-Calvo
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuroinflammation and Centre de Référence Pour les Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM)–Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
- Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Department of Neurology/Neuroimmunology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- * E-mail:
| | - Anne Ruiz
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Chloé Richard
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Sandrine Blondel
- Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Department of Neurology/Neuroimmunology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sylvie Cavagna
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Nathalie Strazielle
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
- BIP Facility, CRNL, Lyon, France
| | - Jean-François Ghersi-Egea
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
- BIP Facility, CRNL, Lyon, France
| | - Pascale Giraudon
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Romain Marignier
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuroinflammation and Centre de Référence Pour les Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM)–Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| |
Collapse
|
64
|
Francisco DMF, Marchetti L, Rodríguez-Lorenzo S, Frías-Anaya E, Figueiredo RM, Winter P, Romero IA, de Vries HE, Engelhardt B, Bruggmann R. Advancing brain barriers RNA sequencing: guidelines from experimental design to publication. Fluids Barriers CNS 2020; 17:51. [PMID: 32811511 PMCID: PMC7433166 DOI: 10.1186/s12987-020-00207-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND RNA sequencing (RNA-Seq) in its varied forms has become an indispensable tool for analyzing differential gene expression and thus characterization of specific tissues. Aiming to understand the brain barriers genetic signature, RNA seq has also been introduced in brain barriers research. This has led to availability of both, bulk and single-cell RNA-Seq datasets over the last few years. If appropriately performed, the RNA-Seq studies provide powerful datasets that allow for significant deepening of knowledge on the molecular mechanisms that establish the brain barriers. However, RNA-Seq studies comprise complex workflows that require to consider many options and variables before, during and after the proper sequencing process. MAIN BODY In the current manuscript, we build on the interdisciplinary experience of the European PhD Training Network BtRAIN ( https://www.btrain-2020.eu/ ) where bioinformaticians and brain barriers researchers collaborated to analyze and establish RNA-Seq datasets on vertebrate brain barriers. The obstacles BtRAIN has identified in this process have been integrated into the present manuscript. It provides guidelines along the entire workflow of brain barriers RNA-Seq studies starting from the overall experimental design to interpretation of results. Focusing on the vertebrate endothelial blood-brain barrier (BBB) and epithelial blood-cerebrospinal-fluid barrier (BCSFB) of the choroid plexus, we provide a step-by-step description of the workflow, highlighting the decisions to be made at each step of the workflow and explaining the strengths and weaknesses of individual choices made. Finally, we propose recommendations for accurate data interpretation and on the information to be included into a publication to ensure appropriate accessibility of the data and reproducibility of the observations by the scientific community. CONCLUSION Next generation transcriptomic profiling of the brain barriers provides a novel resource for understanding the development, function and pathology of these barrier cells, which is essential for understanding CNS homeostasis and disease. Continuous advancement and sophistication of RNA-Seq will require interdisciplinary approaches between brain barrier researchers and bioinformaticians as successfully performed in BtRAIN. The present guidelines are built on the BtRAIN interdisciplinary experience and aim to facilitate collaboration of brain barriers researchers with bioinformaticians to advance RNA-Seq study design in the brain barriers community.
Collapse
Affiliation(s)
- David M F Francisco
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Luca Marchetti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Sabela Rodríguez-Lorenzo
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eduardo Frías-Anaya
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Ricardo M Figueiredo
- GenXPro GmbH, Frankfurt/Main, Germany
- Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | | | - Ignacio Andres Romero
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Helga E de Vries
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland.
| |
Collapse
|
65
|
Litvin DG, Denstaedt SJ, Borkowski LF, Nichols NL, Dick TE, Smith CB, Jacono FJ. Peripheral-to-central immune communication at the area postrema glial-barrier following bleomycin-induced sterile lung injury in adult rats. Brain Behav Immun 2020; 87:610-633. [PMID: 32097765 PMCID: PMC8895345 DOI: 10.1016/j.bbi.2020.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/02/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
The pathways for peripheral-to-central immune communication (P → C I-comm) following sterile lung injury (SLI) are unknown. SLI evokes systemic and central inflammation, which alters central respiratory control and viscerosensory transmission in the nucleus tractus solitarii (nTS). These functional changes coincide with increased interleukin-1 beta (IL-1β) in the area postrema, a sensory circumventricular organ that connects P → C I-comm to brainstem circuits that control homeostasis. We hypothesize that IL-1β and its downstream transcriptional target, cyclooxygenase-2 (COX-2), mediate P → C I-comm in the nTS. In a rodent model of SLI induced by intratracheal bleomycin (Bleo), the sigh frequency and duration of post-sigh apnea increased in Bleo- compared to saline- treated rats one week after injury. This SLI-dependent change in respiratory control occurred concurrently with augmented IL-1β and COX-2 immunoreactivity (IR) in the funiculus separans (FS), a barrier between the AP and the brainstem. At this barrier, increases in IL-1β and COX-2 IR were confined to processes that stained for glial fibrillary acidic protein (GFAP) and that projected basolaterally to the nTS. Further, FS radial-glia did not express TNF-α or IL-6 following SLI. To test our hypothesis, we blocked central COX-1/2 activity by intracerebroventricular (ICV) infusion of Indomethacin (Ind). Continuous ICV Ind treatment prevented Bleo-dependent increases in GFAP + and IL-1β + IR, and restored characteristics of sighs that reset the rhythm. These data indicate that changes in sighs following SLI depend partially on activation of a central COX-dependent P → C I-comm via radial-glia of the FS.
Collapse
Affiliation(s)
- David G Litvin
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Scott J Denstaedt
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Lauren F Borkowski
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO 65212, United States
| | - Nicole L Nichols
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO 65212, United States
| | - Thomas E Dick
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Corey B Smith
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Frank J Jacono
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes VA Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
66
|
Depletion of microglia and macrophages with clodronate liposomes attenuates zymosan-induced Fos expression and hypothermia in the adult mouse. J Neuroimmunol 2020; 344:577244. [DOI: 10.1016/j.jneuroim.2020.577244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/04/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
|
67
|
Eiden LE, Gundlach AL, Grinevich V, Lee MR, Mecawi AS, Chen D, Buijs RM, Hernandez VS, Fajardo-Dolci G, Zhang L. Regulatory peptides and systems biology: A new era of translational and reverse-translational neuroendocrinology. J Neuroendocrinol 2020; 32:e12844. [PMID: 32307768 DOI: 10.1111/jne.12844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
Recently, there has been a resurgence in regulatory peptide science as a result of three converging trends. The first is the increasing population of the drug pipeline with peptide-based therapeutics, mainly in, but not restricted to, incretin-like molecules for treatment of metabolic disorders such as diabetes. The second is the development of genetic and optogenetic tools enabling new insights into how peptides actually function within brain and peripheral circuits to accomplish homeostatic and allostatic regulation. The third is the explosion in defined structures of the G-protein coupled receptors to which most regulatory peptides bind and exert their actions. These trends have closely wedded basic systems biology to drug discovery and development, creating a "two-way street" on which translational advances travel from basic research to the clinic, and, equally importantly, "reverse-translational" information is gathered, about the molecular, cellular and circuit-level mechanisms of action of regulatory peptides, comprising information required for the fine-tuning of drug development through testing in animal models. This review focuses on a small group of 'influential' peptides, including oxytocin, vasopressin, pituitary adenylate cyclase-activating polypeptide, ghrelin, relaxin-3 and glucagon-like peptide-1, and how basic discoveries and their application to therapeutics have intertwined over the past decade.
Collapse
Affiliation(s)
- Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Heath-Intramural Research Program, NIH, Bethesda, MD, USA
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, University Heidelberg, Mannheim, Germany
| | - Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, NIAAA and NIDA, NIH, Bethesda, MD, USA
| | - André S Mecawi
- Laboratory of Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Duan Chen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ruud M Buijs
- Department of Cell Biology and Physiology, Institute for Biomedical Research, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Vito S Hernandez
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Germán Fajardo-Dolci
- School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
68
|
Asami A, Kurganov E, Miyata S. Proliferation of endothelial cells in the choroid plexus of normal and hydrocephalic mice. J Chem Neuroanat 2020; 106:101796. [PMID: 32360474 DOI: 10.1016/j.jchemneu.2020.101796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023]
Abstract
The choroid plexus (CP), located at the walls of the brain ventricles, produces and secretes cerebrospinal fluid (CSF). Hydrocephalus is a neurological disorder in which the CP abnormally secretes excess amounts of CSF into the ventricles. There is currently no information on the vascular dynamics of the CP in adult brains under normal and hydrocephalic conditions. In the present study, we reported the continuous proliferation of endothelial cells in the CP of normal mice, which depended on vascular endothelial cell growth factor (VEGF). The proliferation of endothelial cells increased in mice with intraventricular hemorrhage, which was attenuated by a pretreatment with the toll-like receptor 4 (TLR4) inhibitor VIPER. Moreover, the intracerebroventricular infusion of the TLR4 agonist, lipopolysaccharide, increased endothelial cell proliferation in the CP and induced ventriculomegaly. The present results provide insights into the importance of the TLR4-initiated and VEGF-dependent proliferation of endothelial cells in the pathogenesis of hydrocephalus.
Collapse
Affiliation(s)
- Ayumi Asami
- 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
|
69
|
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: 38] [Impact Index Per Article: 7.6] [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
|
70
|
Silva M, Paiva L, Ratto MH. Ovulation mechanism in South American Camelids: The active role of β-NGF as the chemical signal eliciting ovulation in llamas and alpacas. Theriogenology 2020; 150:280-287. [PMID: 32088046 DOI: 10.1016/j.theriogenology.2020.01.078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 12/22/2022]
Abstract
The ovulation-inducing effect of seminal plasma was first suggested in Bactrian camels over 30 years ago, initiating a long search to identify the 'ovulation-inducing factor' (OIF) present in camelids semen. During the last decade, primarily in llamas and alpacas, this molecule has been intensively studied characterizing its biological and chemical properties and ultimately identifying it as β-Nerve Growth Factor (β-NGF). The high concentration of OIF/β-NGF in seminal plasma of llamas and alpacas, and the striking effects of seminal fluid on ovarian function strongly support the notion of an endocrine mode of action. Also, have challenged the dogma of mating induced ovulation in camelid species, questioning the classical definition of reflex ovulators, which at the light of new evidence should be revised and updated. On the other hand, the presence of OIF/β-NGF and its ovulatory effect in camelids confirm the notion that seminal plasma is not only a transport and survival medium for sperm but also, a signaling agent targeting female tissues after insemination, generating relevant physiological and reproductive consequences. The presence of this molecule, conserved among induced as well as spontaneous ovulating species, clearly suggests that the potential impacts of this reproductive feature extend beyond the camelid species and may have broad implications in mammalian fertility. The aim of the present review is to provide a brief summary of all research efforts undertaken to isolate and identify the ovulation inducing factor present in the seminal plasma of camelids. Also to give an update of the current understanding of the mechanism of action of seminal β-NGF, at central and ovarian level; finally suggesting possible brain targets for this molecule.
Collapse
Affiliation(s)
- Mauricio Silva
- Department of Veterinary Sciences and Public Health, Chile; Nucleus of Research on Agrifood Production, Universidad Católica de Temuco, Temuco, Chile
| | - Luis Paiva
- Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Marcelo H Ratto
- Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
71
|
Gordon L, Blechman J, Shimoni E, Gur D, Anand-Apte B, Levkowitz G. The fenestrae-associated protein Plvap regulates the rate of blood-borne protein passage into the hypophysis. Development 2019; 146:dev.177790. [PMID: 31740533 DOI: 10.1242/dev.177790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
To maintain body homeostasis, endocrine systems must detect and integrate blood-borne peripheral signals. This is mediated by fenestrae, specialized permeable pores in the endothelial membrane. Plasmalemma vesicle-associated protein (Plvap) is located in the fenestral diaphragm and is thought to play a role in the passage of proteins through the fenestrae. However, this suggested function has yet to be demonstrated directly. We studied the development of fenestrated capillaries in the hypophysis, a major neuroendocrine interface between the blood and brain. Using a transgenic biosensor to visualize the vascular excretion of the genetically tagged plasma protein DBP-EGFP, we show that the developmental acquisition of vascular permeability coincides with differential expression of zebrafish plvap orthologs in the hypophysis versus brain. Ultrastructural analysis revealed that plvapb mutants display deficiencies in fenestral diaphragms and increased density of hypophyseal fenestrae. Measurements of DBP-EGFP extravasation in plvapb mutants provided direct proof that Plvap limits the rate of blood-borne protein passage through fenestrated endothelia. We present the regulatory role of Plvap in the development of blood-borne protein detection machinery at a neuroendocrine interface through which hormones are released to the general circulation.
Collapse
Affiliation(s)
- Ludmila Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Eyal Shimoni
- Chemical Research Support, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dvir Gur
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Bela Anand-Apte
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland OH 444195, USA
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| |
Collapse
|
72
|
Korzh V, Kondrychyn I. Origin and development of circumventricular organs in living vertebrate. Semin Cell Dev Biol 2019; 102:13-20. [PMID: 31706729 DOI: 10.1016/j.semcdb.2019.10.010] [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: 08/13/2019] [Accepted: 10/17/2019] [Indexed: 01/22/2023]
Abstract
The circumventricular organs (CVOs) function by mediating chemical communication between blood and brain across the blood-brain barrier. Their origin and developmental mechanisms involved are not understood in enough detail due to a lack of molecular markers common for CVOs. These rather small and inconspicuous organs are found in close vicinity to the third and fourth brain ventricles suggestive of ancient evolutionary origin. Recently, an integrated approach based on analysis of CVOs development in the enhancer-trap transgenic zebrafish led to an idea that almost all of CVOs could be highlighted by GFP expression in this transgenic line. This in turn suggested that an enhancer along with a set of genes it regulates may illustrate the first common element of developmental regulation of CVOs. It seems to be related to a mechanism of suppression of the canonical Wnt/ β-catenin signaling that functions in development of fenestrated capillaries typical for CVOs. Based on that observation the common molecular elements of the putative developmental mechanism of CVOs will be discussed in this review.
Collapse
Affiliation(s)
- Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Poland.
| | | |
Collapse
|
73
|
Lainez NM, Coss D. Obesity, Neuroinflammation, and Reproductive Function. Endocrinology 2019; 160:2719-2736. [PMID: 31513269 PMCID: PMC6806266 DOI: 10.1210/en.2019-00487] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
The increasing occurrence of obesity has become a significant public health concern. Individuals with obesity have higher prevalence of heart disease, stroke, osteoarthritis, diabetes, and reproductive disorders. Reproductive problems include menstrual irregularities, pregnancy complications, and infertility due to anovulation, in women, and lower testosterone and diminished sperm count, in men. In particular, women with obesity have reduced levels of both gonadotropin hormones, and, in obese men, lower testosterone is accompanied by diminished LH. Taken together, these findings indicate central dysregulation of the hypothalamic-pituitary-gonadal axis, specifically at the level of the GnRH neuron function, which is the final brain output for the regulation of reproduction. Obesity is a state of hyperinsulinemia, hyperlipidemia, hyperleptinemia, and chronic inflammation. Herein, we review recent advances in our understanding of how these metabolic and immune changes affect hypothalamic function and regulation of GnRH neurons. In the latter part, we focus on neuroinflammation as a major consequence of obesity and discuss findings that reveal that GnRH neurons are uniquely positioned to respond to inflammatory changes.
Collapse
Affiliation(s)
- Nancy M Lainez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California
- Correspondence: Djurdjica Coss, PhD, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, 303 SOM Research Building, 900 University Avenue, Riverside, California 92521. E-mail:
| |
Collapse
|
74
|
Murayama S, Kurganov E, Miyata S. Activation of microglia and macrophages in the circumventricular organs of the mouse brain during TLR2-induced fever and sickness responses. J Neuroimmunol 2019; 334:576973. [PMID: 31170673 DOI: 10.1016/j.jneuroim.2019.576973] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/26/2019] [Accepted: 05/27/2019] [Indexed: 01/28/2023]
Abstract
Toll-like receptor 2 (TLR2) recognizes cell wall components from Gram-positive bacteria. Until now, however, little has been known about the significance of brain TLR2 in controlling inflammation and thermoregulatory responses during systemic Gram-positive bacterial infection. In the present study, the TLR2 immunoreactivity was seen to be prominent in the microglia/macrophages of the circumventricular organs (CVOs) of the mouse brain. The intraperitoneal injection of Pam3CSK4, a TLR2 agonist, induced nuclear factor-κ B activation in the microglia/macrophages of the CVOs. The injection of Pam3CSK4 also produced the expression of Fos at astrocytes and neurons in the CVOs and the regions neighboring the CVOs. The Pam3CSK4 injection induced fever and sickness responses. Pretreatment with lipopolysaccharide, a TLR4 agonist, augmented the Pam3CSK4-induced fever together with the increased TLR2 immunoreactivity. These results indicate that the TLR2 in microglia/macrophages of the CVOs are possibly associated with initiating and transmitting inflammatory responses in the brain.
Collapse
Affiliation(s)
- Saki Murayama
- 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
|
75
|
Zielinski MR, Systrom DM, Rose NR. Fatigue, Sleep, and Autoimmune and Related Disorders. Front Immunol 2019; 10:1827. [PMID: 31447842 PMCID: PMC6691096 DOI: 10.3389/fimmu.2019.01827] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022] Open
Abstract
Profound and debilitating fatigue is the most common complaint reported among individuals with autoimmune disease, such as systemic lupus erythematosus, multiple sclerosis, type 1 diabetes, celiac disease, chronic fatigue syndrome, and rheumatoid arthritis. Fatigue is multi-faceted and broadly defined, which makes understanding the cause of its manifestations especially difficult in conditions with diverse pathology including autoimmune diseases. In general, fatigue is defined by debilitating periods of exhaustion that interfere with normal activities. The severity and duration of fatigue episodes vary, but fatigue can cause difficulty for even simple tasks like climbing stairs or crossing the room. The exact mechanisms of fatigue are not well-understood, perhaps due to its broad definition. Nevertheless, physiological processes known to play a role in fatigue include oxygen/nutrient supply, metabolism, mood, motivation, and sleepiness-all which are affected by inflammation. Additionally, an important contributing element to fatigue is the central nervous system-a region impacted either directly or indirectly in numerous autoimmune and related disorders. This review describes how inflammation and the central nervous system contribute to fatigue and suggests potential mechanisms involved in fatigue that are likely exhibited in autoimmune and related diseases.
Collapse
Affiliation(s)
- Mark R Zielinski
- Veterans Affairs Boston Healthcare System, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - David M Systrom
- Department of Medicine, Harvard Medical School, Boston, MA, United States.,Department of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Noel R Rose
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
76
|
Wickham RJ. Revisiting the physiology of nausea and vomiting-challenging the paradigm. Support Care Cancer 2019; 28:13-21. [PMID: 31388745 DOI: 10.1007/s00520-019-05012-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE The predominant neurotransmitters and receptors for acute and delayed chemotherapy-induced nausea and vomiting (CINV) are represented in the current paradigm, which reflects successful control of emesis. However, control of nausea (N) lags behind management of vomiting (V). This review aims to re-examine and incorporate new information about the mechanisms of V and N. METHODS The initial literature search focused on CINV. Keywords in articles led to subsequent discovery of publications focused on N&V in other medical and scientific fields (e.g., gastroenterology, neurology, cannabinoid science, neuropharmacology, and motion sickness). Using keywords to identify other sources continued until no further recent, meaningful publications were found. RESULTS More than 86% of references were from recent non-oncology journals and books, suggesting there are many areas for cross-fertilization research into mechanisms and management of N&V-particularly of N, which involves overlapping and dissimilar CNS areas from V. Information from cited articles was incorporated into visual representation of N&V, which is certainly not exhaustive but supports highly complex processes in the stomach and gut, the vagus nerve and spinal cord neurons, the nucleus tractus solitarii, and the anterior insular cortex and anterior cingulate cortex with input from the amygdala. CONCLUSIONS These data support the idea that mechanisms for N, whatever the cause, must be highly similar. Continued research into nausea, including patient-reported evaluation and outcomes, is important; interventions for nausea could be considered adjuvants to current standard of care antiemetics and be individualized, depending on patient-reported efficacy and adverse effects and preferences.
Collapse
Affiliation(s)
- Rita J Wickham
- Rush University College of Nursing, 8039 Garth Point Lane, Rapid River, MI, 49878, USA.
| |
Collapse
|
77
|
Pyrosequencing versus methylation-specific PCR for assessment of MGMT methylation in tumor and blood samples of glioblastoma patients. Sci Rep 2019; 9:11125. [PMID: 31366977 PMCID: PMC6668570 DOI: 10.1038/s41598-019-47642-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
Circulating biomarkers in blood may provide an interesting alternative to risky tissue biopsies in the diagnosis and follow-up of glioblastoma patients. We have assessed MGMT methylation status in blood and tissue samples from unresected glioblastoma patients who had been included in the randomized GENOM-009 trial. Paired blood and tissue samples were assessed by methylation-specific PCR (MSP) and pyrosequencing (PYR). After establishing the minimum PYR cut-off that could yield a significant difference in overall survival, we assessed the sensitivity, specificity, positive predictive value and negative predictive value (NPV) of the analyses. Methylation could be detected in cfDNA by both MSP and PYR but with low concordance with results in tissue. Sensitivity was low for both methods (31% and 38%, respectively), while specificity was higher for MSP in blood than for PYR in plasma (96% vs 76%) and NPV was similar (56 vs 57%). Concordance of results in tissue by MSP and PYR was 84.3% (P < 0.001) and correlated with outcome. We conclude that detection of cfDNA in the blood of glioblastoma patients can be an alternative when tumor tissue is not available but methods for the detection of cfDNA in blood must improve before it can replace analysis in tumor tissue.
Collapse
|
78
|
Hiratsuka D, Kurganov E, Furube E, Morita M, Miyata S. VEGF- and PDGF-dependent proliferation of oligodendrocyte progenitor cells in the medulla oblongata after LPC-induced focal demyelination. J Neuroimmunol 2019; 332:176-186. [DOI: 10.1016/j.jneuroim.2019.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 12/27/2022]
|
79
|
Oral gavage of capsaicin causes TRPV1-dependent acute hypothermia and TRPV1-independent long-lasting increase of locomotor activity in the mouse. Physiol Behav 2019; 206:213-224. [DOI: 10.1016/j.physbeh.2019.04.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/20/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022]
|
80
|
Perello M, Cabral A, Cornejo MP, De Francesco PN, Fernandez G, Uriarte M. Brain accessibility delineates the central effects of circulating ghrelin. J Neuroendocrinol 2019; 31:e12677. [PMID: 30582239 DOI: 10.1111/jne.12677] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Ghrelin is a hormone produced in the gastrointestinal tract that acts via the growth hormone secretagogue receptor. In the central nervous system, ghrelin signalling is able to recruit different neuronal targets that regulate the behavioural, neuroendocrine, metabolic and autonomic effects of the hormone. Notably, several studies using radioactive or fluorescent variants of ghrelin have found that the accessibility of circulating ghrelin into the mouse brain is both strikingly low and restricted to some specific brain areas. A variety of studies addressing central effects of systemically injected ghrelin in mice have also provided indirect evidence that the accessibility of plasma ghrelin into the brain is limited. Here, we review these previous observations and discuss the putative pathways that would allow plasma ghrelin to gain access into the brain together with their physiological implications. Additionally, we discuss some potential features regarding the accessibility of plasma ghrelin into the human brain based on the observations reported by studies that investigate the consequences of ghrelin administration to humans.
Collapse
Affiliation(s)
- Mario Perello
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Cabral
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - María P Cornejo
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Maia Uriarte
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
81
|
Muneoka S, Murayama S, Nakano Y, Miyata S. TLR4 in circumventricular neural stem cells is a negative regulator for thermogenic pathways in the mouse brain. J Neuroimmunol 2019; 331:58-73. [DOI: 10.1016/j.jneuroim.2018.04.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/23/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022]
|
82
|
Shell B, Farmer GE, Nedungadi TP, Wang LA, Marciante AB, Snyder B, Cunningham RL, Cunningham JT. Angiotensin type 1a receptors in the median preoptic nucleus support intermittent hypoxia-induced hypertension. Am J Physiol Regul Integr Comp Physiol 2019; 316:R651-R665. [PMID: 30892911 PMCID: PMC6589598 DOI: 10.1152/ajpregu.00393.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/13/2019] [Indexed: 01/19/2023]
Abstract
Chronic intermittent hypoxia (CIH) is a model of the hypoxemia from sleep apnea that causes a sustained increase in blood pressure. Inhibition of the central renin-angiotensin system or FosB in the median preoptic nucleus (MnPO) prevents the sustained hypertensive response to CIH. We tested the hypothesis that angiotensin type 1a (AT1a) receptors in the MnPO, which are upregulated by CIH, contribute to this hypertension. In preliminary experiments, retrograde tract tracing studies showed AT1a receptor expression in MnPO neurons projecting to the paraventricular nucleus. Adult male rats were exposed to 7 days of intermittent hypoxia (cycling between 21% and 10% O2 every 6 min, 8 h/day during light phase). Seven days of CIH was associated with a FosB-dependent increase in AT1a receptor mRNA without changes in the permeability of the blood-brain barrier in the MnPO. Separate groups of rats were injected in the MnPO with an adeno-associated virus containing short hairpin (sh)RNA against AT1a receptors to test their role in intermittent hypoxia hypertension. Injections of shRNA against AT1a in MnPO blocked the increase in mRNA associated with CIH, prevented the sustained component of the hypertension during normoxia, and reduced circulating advanced oxidation protein products, an indicator of oxidative stress. Rats injected with shRNA against AT1a and exposed to CIH had less FosB staining in MnPO and the rostral ventrolateral medulla after intermittent hypoxia than rats injected with the control vector that were exposed to CIH. Our results indicate AT1a receptors in the MnPO contribute to the sustained blood pressure increase to intermittent hypoxia.
Collapse
MESH Headings
- Angiotensin II/administration & dosage
- Animals
- Blood Pressure/drug effects
- Disease Models, Animal
- Hypertension/etiology
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Hypoxia/complications
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia/physiopathology
- Injections, Intraventricular
- Male
- Oxidative Stress
- Preoptic Area/drug effects
- Preoptic Area/metabolism
- Preoptic Area/physiopathology
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Signal Transduction
- Up-Regulation
Collapse
Affiliation(s)
- Brent Shell
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - T Prashant Nedungadi
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Lei A Wang
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Alexandria B Marciante
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Brina Snyder
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
83
|
Dos-Santos RC, Reis LC, Perello M, Ferguson AV, Mecawi AS. The actions of ghrelin in the paraventricular nucleus: energy balance and neuroendocrine implications. Ann N Y Acad Sci 2019; 1455:81-97. [PMID: 31008525 DOI: 10.1111/nyas.14087] [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: 12/21/2018] [Revised: 02/28/2019] [Accepted: 03/10/2019] [Indexed: 12/15/2022]
Abstract
Ghrelin is a peptide mainly produced and secreted by the stomach. Since its discovery, the impact of ghrelin on the regulation of food intake has been the most studied function of this hormone; however, ghrelin affects a wide range of physiological systems, many of which are controlled by the hypothalamic paraventricular nucleus (PVN). Several pathways may mediate the effects of ghrelin on PVN neurons, such as direct or indirect effects mediated by circumventricular organs and/or the arcuate nucleus. The ghrelin receptor is expressed in PVN neurons, and the peripheral or intracerebroventricular administration of ghrelin affects PVN neuronal activity. Intra-PVN application of ghrelin increases food intake and decreases fat oxidation, which chronically contribute to the increased adiposity. Additionally, ghrelin modulates the neuroendocrine axes controlled by the PVN, increasing the release of vasopressin and oxytocin by magnocellular neurons and corticotropin-releasing hormone by neuroendocrine parvocellular neurons, while possibly inhibiting the release of thyrotropin-releasing hormone. Thus, the PVN is an important target for the actions of ghrelin. Our review discusses the mechanisms of ghrelin actions in the PVN, and its potential implications for energy balance, neuroendocrine, and integrative physiological control.
Collapse
Affiliation(s)
- Raoni C Dos-Santos
- Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Luís C Reis
- Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology, La Plata, Argentina
| | - Alastair V Ferguson
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - André S Mecawi
- Laboratory of Neuroendocrinology, Department of Biophysics, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
84
|
Sou K, Le DL, Sato H. Nanocapsules for Programmed Neurotransmitter Release: Toward Artificial Extracellular Synaptic Vesicles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900132. [PMID: 30887709 DOI: 10.1002/smll.201900132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/20/2019] [Indexed: 06/09/2023]
Abstract
Nanocapsules present a promising platform for delivering chemicals and biomolecules to a site of action in a living organism. Because the biological action of the encapsulated molecules is blocked until they are released from the nanocapsules, the encapsulation structure enables triggering of the topical and timely action of the molecules at the target site. A similar mechanism seems promising for the spatiotemporal control of signal transduction triggered by the release of signal molecules in neuronal, metabolic, and immune systems. From this perspective, nanocapsules can be regarded as practical tools to apply signal molecules such as neurotransmitters to intervene in signal transduction. However, spatiotemporal control of the payload release from nanocapsules persists as a key technical issue. Stimulus-responsive nanocapsules that release payloads in response to external input of physical stimuli are promising platforms to enable programmed payload release. These programmable nanocapsules encapsulating neurotransmitters are expected to lead to new insights and perspectives related to artificial extracellular synaptic vesicles that might provide an experimental and therapeutic strategy for neuromodulation and nervous system disorders.
Collapse
Affiliation(s)
- Keitaro Sou
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku, Tokyo, 169-8555, Japan
| | - Duc Long Le
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Hirotaka Sato
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
85
|
Yoo S, Cha D, Kim DW, Hoang TV, Blackshaw S. Tanycyte-Independent Control of Hypothalamic Leptin Signaling. Front Neurosci 2019; 13:240. [PMID: 30941008 PMCID: PMC6433882 DOI: 10.3389/fnins.2019.00240] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/01/2019] [Indexed: 12/29/2022] Open
Abstract
Leptin is secreted by adipocytes to regulate appetite and body weight. Recent studies have reported that tanycytes actively transport circulating leptin across the brain barrier into the hypothalamus, and are required for normal levels of hypothalamic leptin signaling. However, direct evidence for leptin receptor (LepR) expression is lacking, and the effect of tanycyte-specific deletion of LepR has not been investigated. In this study, we analyze the expression and function of the tanycytic LepR in mice. Using single-molecule fluorescent in situ hybridization (smfISH), RT-qPCR, single-cell RNA sequencing (scRNA-Seq), and selective deletion of the LepR in tanycytes, we are unable to detect expression of LepR in the tanycytes. Tanycyte-specific deletion of LepR likewise did not affect leptin-induced pSTAT3 expression in hypothalamic neurons, regardless of whether leptin was delivered by intraperitoneal or intracerebroventricular injection. Finally, we use activity-regulated scRNA-Seq (act-Seq) to comprehensively profile leptin-induced changes in gene expression in all cell types in mediobasal hypothalamus. Clear evidence for leptin signaling is only seen in endothelial cells and subsets of neurons, although virtually all cell types show leptin-induced changes in gene expression. We thus conclude that LepR expression in tanycytes is either absent or undetectably low, that tanycytes do not directly regulate hypothalamic leptin signaling through a LepR-dependent mechanism, and that leptin regulates gene expression in diverse hypothalamic cell types through both direct and indirect mechanisms.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - David Cha
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States.,Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, Johns Hopkins University, Baltimore, MD, United States.,Center for Human Systems Biology, Johns Hopkins University, Baltimore, MD, United States.,School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
86
|
Hillebrand S, Schanda K, Nigritinou M, Tsymala I, Böhm D, Peschl P, Takai Y, Fujihara K, Nakashima I, Misu T, Reindl M, Lassmann H, Bradl M. Circulating AQP4-specific auto-antibodies alone can induce neuromyelitis optica spectrum disorder in the rat. Acta Neuropathol 2019; 137:467-485. [PMID: 30564980 PMCID: PMC6514074 DOI: 10.1007/s00401-018-1950-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022]
Abstract
It is well established that the binding of pathogenic aquaporin-4 (AQP4)-specific autoantibodies to astrocytes may initiate a cascade of events culminating in the destruction of these cells and in the formation of large tissue-destructive lesions typical for patients with neuromyelitis optica spectrum disorders (NMOSD). To date, not a single experimental study has shown that the systemic presence of the antibody alone can induce any damage to the central nervous system (CNS), while pathological studies on brains of NMOSD patients suggested that there might be ways for antibody entry and subsequent tissue damage. Here, we systemically applied a highly pathogenic, monoclonal antibody with high affinity to AQP4 over prolonged period of time to rats, and show that AQP4-abs can enter the CNS on their own, via circumventricular organs and meningeal or parenchymal blood vessels, that these antibodies initiate the formation of radically different lesions with AQP4 loss, depending on their mode and site of entry, and that lesion formation is much more efficient in the presence of encephalitogenic T-cell responses. We further demonstrate that the established tissue-destructive lesions trigger the formation of additional lesions by short and far reaching effects on blood vessels and their branches, and that AQP4-abs have profound effects on the AQP4 expression in peripheral tissues which counter-act possible titer loss by antibody absorption outside the CNS. Cumulatively, these data indicate that directly induced pathological changes caused by AQP4-abs inside and outside the CNS are efficient drivers of disease evolution in seropositive organisms.
Collapse
Affiliation(s)
- Sophie Hillebrand
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Kathrin Schanda
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Magdalini Nigritinou
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Irina Tsymala
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Denise Böhm
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Patrick Peschl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Yoshiki Takai
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuo Fujihara
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ichiro Nakashima
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Markus Reindl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
87
|
Schier LA, Spector AC. The Functional and Neurobiological Properties of Bad Taste. Physiol Rev 2019; 99:605-663. [PMID: 30475657 PMCID: PMC6442928 DOI: 10.1152/physrev.00044.2017] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/18/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022] Open
Abstract
The gustatory system serves as a critical line of defense against ingesting harmful substances. Technological advances have fostered the characterization of peripheral receptors and have created opportunities for more selective manipulations of the nervous system, yet the neurobiological mechanisms underlying taste-based avoidance and aversion remain poorly understood. One conceptual obstacle stems from a lack of recognition that taste signals subserve several behavioral and physiological functions which likely engage partially segregated neural circuits. Moreover, although the gustatory system evolved to respond expediently to broad classes of biologically relevant chemicals, innate repertoires are often not in register with the actual consequences of a food. The mammalian brain exhibits tremendous flexibility; responses to taste can be modified in a specific manner according to bodily needs and the learned consequences of ingestion. Therefore, experimental strategies that distinguish between the functional properties of various taste-guided behaviors and link them to specific neural circuits need to be applied. Given the close relationship between the gustatory and visceroceptive systems, a full reckoning of the neural architecture of bad taste requires an understanding of how these respective sensory signals are integrated in the brain.
Collapse
Affiliation(s)
- Lindsey A Schier
- Department of Biological Sciences, University of Southern California , Los Angeles, California ; and Department of Psychology and Program in Neuroscience, Florida State University , Tallahassee, Florida
| | - Alan C Spector
- Department of Biological Sciences, University of Southern California , Los Angeles, California ; and Department of Psychology and Program in Neuroscience, Florida State University , Tallahassee, Florida
| |
Collapse
|
88
|
Lainez NM, Coss D. Leukemia Inhibitory Factor Represses GnRH Gene Expression via cFOS during Inflammation in Male Mice. Neuroendocrinology 2019; 108:291-307. [PMID: 30630179 PMCID: PMC6561803 DOI: 10.1159/000496754] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanisms whereby neuroinflammation negatively affects neuronal function in the hypothalamus are not clear. Our previous study determined that obesity-mediated chronic inflammation elicits sex-specific impairment in reproductive function via reduction in spine density in gonadotropin-releasing hormone (GnRH) neurons. Neuroinflammation and subsequent decrease in GnRH neuron spine density was specific for male mice, while protection in females was independent of ovarian estrogens. METHODS To examine if neuroinflammation-induced cytokines can directly regulate GnRH gene expression, herein we examined signaling pathways and mechanisms in males in vivo and in GnRH-expressing cell line, GT1-7. RESULTS GnRH neurons express cytokine receptors, and chronic or acute neuroinflammation represses GnRH gene expression in vivo. Leukemia inhibitory factor (LIF) in particular represses GnRH expression in GT1-7 cells, while other cytokines do not. STAT3 and MAPK pathways are activated following LIF treatment, but only MAPK pathway, specifically p38α, is sufficient to repress the GnRH gene. LIF induces cFOS that represses the GnRH gene via the -1,793 site in the enhancer region. In vivo, following high-fat diet, cFOS is induced in GnRH neurons and neurons juxtaposed to the leaky blood brain barrier of the organum vasculosum of the lamina terminalis, but not in the neurons further away. CONCLUSION Our results indicate that the increase in LIF due to neuroinflammation induces cFOS and represses the GnRH gene. Therefore, in addition to synaptic changes in GnRH neurons, neuroinflammatory cytokines directly regulate gene expression and reproductive function, and the specificity for neuronal targets may stem from the proximity to the fenestrated capillaries.
Collapse
Affiliation(s)
- Nancy M Lainez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA,
| |
Collapse
|
89
|
Bentivoglio M, Kristensson K, Rottenberg ME. Circumventricular Organs and Parasite Neurotropism: Neglected Gates to the Brain? Front Immunol 2018; 9:2877. [PMID: 30619260 PMCID: PMC6302769 DOI: 10.3389/fimmu.2018.02877] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/22/2018] [Indexed: 12/20/2022] Open
Abstract
Circumventricular organs (CVOs), neural structures located around the third and fourth ventricles, harbor, similarly to the choroid plexus, vessels devoid of a blood-brain barrier (BBB). This enables them to sense immune-stimulatory molecules in the blood circulation, but may also increase chances of exposure to microbes. In spite of this, attacks to CVOs by microbes are rarely described. It is here highlighted that CVOs and choroid plexus can be infected by pathogens circulating in the bloodstream, providing a route for brain penetration, as shown by infections with the parasites Trypanosoma brucei. Immune responses elicited by pathogens or systemic infections in the choroid plexus and CVOs are briefly outlined. From the choroid plexus trypanosomes can seed into the ventricles and initiate accelerated infiltration of T cells and parasites in periventricular areas. The highly motile trypanosomes may also enter the brain parenchyma from the median eminence, a CVO located at the base of the third ventricle, by crossing the border into the BBB-protected hypothalamic arcuate nuclei. A gate may, thus, be provided for trypanosomes to move into brain areas connected to networks of regulation of circadian rhythms and sleep-wakefulness, to which other CVOs are also connected. Functional imbalances in these networks characterize human African trypanosomiasis, also called sleeping sickness. They are distinct from the sickness response to bacterial infections, but can occur in common neuropsychiatric diseases. Altogether the findings lead to the question: does the neglect in reporting microbe attacks to CVOs reflect lack of awareness in investigations or of gate-opening capability by microbes?
Collapse
Affiliation(s)
- Marina Bentivoglio
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Martin E. Rottenberg
- Department Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
90
|
Anbalagan S, Gordon L, Blechman J, Matsuoka RL, Rajamannar P, Wircer E, Biran J, Reuveny A, Leshkowitz D, Stainier DYR, Levkowitz G. Pituicyte Cues Regulate the Development of Permeable Neuro-Vascular Interfaces. Dev Cell 2018; 47:711-726.e5. [PMID: 30449506 DOI: 10.1016/j.devcel.2018.10.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/07/2018] [Accepted: 10/15/2018] [Indexed: 11/30/2022]
Abstract
The hypothalamo-neurohypophyseal system (HNS) regulates homeostasis through the passage of neurohormones and blood-borne proteins via permeable blood capillaries that lack the blood-brain barrier (BBB). Why neurohypophyseal capillaries become permeable while the neighboring vasculature of the brain forms BBB remains unclear. We show that pituicytes, the resident astroglial cells of the neurohypophysis, express genes that are associated with BBB breakdown during neuroinflammation. Pituicyte-enriched factors provide a local microenvironment that instructs a permeable neurovascular conduit. Thus, genetic and pharmacological perturbations of Vegfa and Tgfβ3 affected HNS vascular morphogenesis and permeability and impaired the expression of the fenestral marker plvap. The anti-inflammatory agent dexamethasone decreased HNS permeability and downregulated the pituicyte-specific cyp26b gene, encoding a retinoic acid catabolic enzyme. Inhibition of Cyp26b activity led to upregulation of tight junction protein Claudin-5 and decreased permeability. We conclude that pituicyte-derived factors regulate the "decision" of endothelial cells to adopt a permeable endothelial fate instead of forming a BBB.
Collapse
Affiliation(s)
- Savani Anbalagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Ludmila Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Ryota L Matsuoka
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Preethi Rajamannar
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Einav Wircer
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Jakob Biran
- Department of Poultry and Aquaculture, Agricultural Research Organization, Rishon Letziyon 7528809, Israel
| | - Adriana Reuveny
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Bioinformatics Unit, LSCF Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|
91
|
Zeiner PS, Brandhofe A, Müller-Eschner M, Steinmetz H, Pfeilschifter W. Area postrema syndrome as frequent feature of Bickerstaff brainstem encephalitis. Ann Clin Transl Neurol 2018; 5:1534-1542. [PMID: 30564620 PMCID: PMC6292382 DOI: 10.1002/acn3.666] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/27/2018] [Accepted: 09/10/2018] [Indexed: 01/12/2023] Open
Abstract
Objective Area postrema (AP) syndrome (defined as: nausea and/or emesis and/or singultus at onset of brainstem dysfunction) comprises complex pathophysiologic mechanisms triggered by different entities. The first objective was to assess the frequency of AP syndrome as a clinical feature in brainstem encephalitis (BE). Finding an especially high prevalence of AP syndrome in Bickerstaff brainstem encephalitis (BBE), we also analyzed the frequency of AP syndrome in other autoimmune diseases with anti-ganglioside antibodies (Guillain-Barré syndrome (GBS) and its variants). Methods We systematically evaluated the prevalence of AP syndrome in BE in all patients treated at our university hospital during a 15-year period. In a second step, BBE patients were compared to GBS and Miller Fisher syndrome (MFS) patients as clinical subtypes of a disease continuum without brainstem dysfunction. Results We found AP syndrome in 8 of 21 BE patients, including 3 of 7 BBE and in 4 of 112 GBS/MFS patients. AP syndrome was as a frequent but under-recognized feature of BE with a significant impact on patients' well being. Interpretation Manifestation of AP syndrome in BBE but also in GBS and its subtypes point toward a role of autoimmune antibodies that should be investigated in future studies. Considerable misdiagnosis or nonrecognition complicates diagnostic and therapeutic management. Therefore, AP syndrome should be considered in any episode of otherwise unexplained nausea, emesis, or singultus.
Collapse
Affiliation(s)
- Pia S Zeiner
- Department of Neurology University Hospital/Goethe University Frankfurt Schleusenweg 2-16 Frankfurt/Main 60528 Germany.,Dr. Senckenberg Institute of Neurooncology University Hospital/Goethe University Frankfurt Heinrich-Hoffmann-Strasse 7 Frankfurt/Main 60528 Germany
| | - Annemarie Brandhofe
- Department of Neurology University Hospital/Goethe University Frankfurt Schleusenweg 2-16 Frankfurt/Main 60528 Germany
| | - Monika Müller-Eschner
- Institute of Neuroradiology University Hospital/Goethe University Frankfurt Schleusenweg 2-16 Frankfurt/Main 60528 Germany
| | - Helmuth Steinmetz
- Department of Neurology University Hospital/Goethe University Frankfurt Schleusenweg 2-16 Frankfurt/Main 60528 Germany
| | - Waltraud Pfeilschifter
- Department of Neurology University Hospital/Goethe University Frankfurt Schleusenweg 2-16 Frankfurt/Main 60528 Germany
| |
Collapse
|
92
|
Ludewig P, Winneberger J, Magnus T. The cerebral endothelial cell as a key regulator of inflammatory processes in sterile inflammation. J Neuroimmunol 2018; 326:38-44. [PMID: 30472304 DOI: 10.1016/j.jneuroim.2018.10.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/17/2018] [Accepted: 10/24/2018] [Indexed: 11/18/2022]
Abstract
Cerebral endothelial cells accomplish numerous tasks connected to the maintenance of homeostasis of the central nervous system. They create a barrier between the central nervous system and peripheral blood and regulate mechanotransduction, vascular permeability, rheology, thrombogenesis, and leukocyte adhesion. In pathophysiological conditions (e.g., stroke or ischemia-reperfusion injury) the endothelial functions are impaired, leading to increased vascular permeability, vascular inflammation, leukocyte-endothelium interactions, and transendothelial migration, driving CNS inflammation and neuronal destruction. This review describes the current knowledge on the regulatory roles of endothelial cells in neuroinflammatory processes.
Collapse
Affiliation(s)
- Peter Ludewig
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany.
| | - Jack Winneberger
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| |
Collapse
|
93
|
Morris G, Fernandes BS, Puri BK, Walker AJ, Carvalho AF, Berk M. Leaky brain in neurological and psychiatric disorders: Drivers and consequences. Aust N Z J Psychiatry 2018; 52:924-948. [PMID: 30231628 DOI: 10.1177/0004867418796955] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The blood-brain barrier acts as a highly regulated interface; its dysfunction may exacerbate, and perhaps initiate, neurological and neuropsychiatric disorders. METHODS In this narrative review, focussing on redox, inflammatory and mitochondrial pathways and their effects on the blood-brain barrier, a model is proposed detailing mechanisms which might explain how increases in blood-brain barrier permeability occur and can be maintained with increasing inflammatory and oxidative and nitrosative stress being the initial drivers. RESULTS Peripheral inflammation, which is causatively implicated in the pathogenesis of major psychiatric disorders, is associated with elevated peripheral pro-inflammatory cytokines, which in turn cause increased blood-brain barrier permeability. Reactive oxygen species, such as superoxide radicals and hydrogen peroxide, and reactive nitrogen species, such as nitric oxide and peroxynitrite, play essential roles in normal brain capillary endothelial cell functioning; however, chronically elevated oxidative and nitrosative stress can lead to mitochondrial dysfunction and damage to the blood-brain barrier. Activated microglia, redox control of which is mediated by nitric oxide synthases and nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, secrete neurotoxic molecules such as reactive oxygen species, nitric oxide, prostaglandin, cyclooxygenase-2, quinolinic acid, several chemokines (including monocyte chemoattractant protein-1 [MCP-1], C-X-C motif chemokine ligand 1 [CXCL-1] and macrophage inflammatory protein 1α [MIP-1α]) and the pro-inflammatory cytokines interleukin-6, tumour necrosis factor-α and interleukin-1β, which can exert a detrimental effect on blood-brain barrier integrity and function. Similarly, reactive astrocytes produce neurotoxic molecules such as prostaglandin E2 and pro-inflammatory cytokines, which can cause a 'leaky brain'. CONCLUSION Chronic inflammatory and oxidative and nitrosative stress is associated with the development of a 'leaky gut'. The following evidence-based approaches, which address the leaky gut and blood-brain barrier dysfunction, are suggested as potential therapeutic interventions for neurological and neuropsychiatric disorders: melatonin, statins, probiotics containing Bifidobacteria and Lactobacilli, N-acetylcysteine, and prebiotics containing fructo-oligosaccharides and galacto-oligosaccharides.
Collapse
Affiliation(s)
- Gerwyn Morris
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia
| | - Brisa S Fernandes
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia.,2 Centre for Addiction and Mental Health (CAMH) and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Basant K Puri
- 3 Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Adam J Walker
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia
| | - Andre F Carvalho
- 2 Centre for Addiction and Mental Health (CAMH) and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Michael Berk
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia.,4 Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
94
|
Abstract
Astrocytes have historically been considered structural supporting cells for neurons. Thanks to new molecular tools, allowing specific cell ablation or over-expression of genes, new unexpected astrocytic functions have recently been unveiled. This review focus on emerging groundbreaking findings showing that hypothalamic astrocytes are pivotal for the regulation of whole body energy homeostasis. Hypothalamic astrocytes sense glucose and fatty acids, and express receptors for several peripheral hormones such as leptin and insulin. Furthermore, they display striking sexual dimorphism which may account, at least partially, for gender specific differences in energy homeostasis. Metabolic alterations have been shown to influence the initiation and progression of many neurodegenerative disorders. A better understanding of the roles and interplay between the different brain cells in regulating energy homeostasis could help develop new therapeutic strategies to prevent or cure neurodegenerative disorders.
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
95
|
Lainez NM, Jonak CR, Nair MG, Ethell IM, Wilson EH, Carson MJ, Coss D. Diet-Induced Obesity Elicits Macrophage Infiltration and Reduction in Spine Density in the Hypothalami of Male but Not Female Mice. Front Immunol 2018; 9:1992. [PMID: 30254630 PMCID: PMC6141693 DOI: 10.3389/fimmu.2018.01992] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/13/2018] [Indexed: 01/23/2023] Open
Abstract
Increasing prevalence in obesity has become a significant public concern. C57BL/6J mice are prone to diet-induced obesity (DIO) when fed high-fat diet (HFD), and develop chronic inflammation and metabolic syndrome, making them a good model to analyze mechanisms whereby obesity elicits pathologies. DIO mice demonstrated profound sex differences in response to HFD with respect to inflammation and hypothalamic function. First, we determined that males are prone to DIO, while females are resistant. Ovariectomized females, on the other hand, are susceptible to DIO, implying protection by ovarian hormones. Males, but not females, exhibit changes in hypothalamic neuropeptide expression. Surprisingly, ovariectomized females remain resistant to neuroendocrine changes, showing that ovarian hormones are not necessary for protection. Second, obese mice exhibit sex differences in DIO-induced inflammation. Microglial activation and peripheral macrophage infiltration is seen in the hypothalami of males, while females are protected from the increase in inflammatory cytokines and do not exhibit microglia morphology changes nor monocyte-derived macrophage infiltration, regardless of the presence of ovarian hormones. Strikingly, the anti-inflammatory cytokine IL-10 is increased in the hypothalami of females but not males. Third, this study posits a potential mechanism of obesity-induced impairment of hypothalamic function whereby obese males exhibit reduced levels of synaptic proteins in the hypothalamus and fewer spines in GnRH neurons, located in the areas exhibiting macrophage infiltration. Our studies suggest that inflammation-induced synaptic remodeling is potentially responsible for hypothalamic impairment that may contribute to diminished levels of gonadotropin hormones, testosterone, and sperm numbers, which we observe and corresponds to the observations in obese humans. Taken together, our data implicate neuro-immune mechanisms underlying sex-specific differences in obesity-induced impairment of the hypothalamic function with potential consequences for reproduction and fertility.
Collapse
Affiliation(s)
- Nancy M Lainez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Monica J Carson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
96
|
Jiang R, Prell C, Lönnerdal B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life. FASEB J 2018; 33:1681-1694. [PMID: 30199283 DOI: 10.1096/fj.201701290rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Osteopontin (OPN) is a pleiotropic protein and is abundantly present in milk. Its functions include immune modulation and cellular proliferation and differentiation. OPN is highly expressed in the brain. We investigated the effects of milk-derived OPN on brain development of mouse pups. Wild-type (WT) dams producing OPN+ milk and OPN knockout (KO) dams producing OPN- milk nursed WT pups (OPN+/+), yielding 2 pup treatment groups, OPN+ OPN+/+ and OPN- OPN+/+, for comparison. Preliminary studies supported use of this model by showing high concentrations of OPN in milk of WT dams and no OPN in milk of OPN KO dams, and production of similar amounts of milk by WT and KO dams. The ability of ingested milk OPN to enter the brain was revealed by appearance of orally gavaged [125I]-labeled and antibody-probed milk OPN in brains of pups. Brain OPN mRNA levels were similar in both nursed groups, but the brain OPN protein level was significantly lower in the OPN- OPN+/+ group at postnatal days 6 and 8. Behavior tests showed impaired memory and learning ability in OPN- OPN+/+ pups. In addition, our study revealed increased expression of myelination-related proteins and elevated proliferation and differentiation of NG-2 glia into oligodendrocytes in the brain of OPN+ OPN+/+ pups, accompanied by increased activation of ERK-1/2 and PI3K/Akt signaling. We concluded that milk OPN can play an important role in brain development and behavior in infancy by promoting myelination.-Jiang, R., Prell, C., Lönnerdal, B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life.
Collapse
Affiliation(s)
- Rulan Jiang
- Department of Nutrition, University of California Davis, Davis, California, USA
| | - Christine Prell
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Bo Lönnerdal
- Department of Nutrition, University of California Davis, Davis, California, USA
| |
Collapse
|
97
|
Al-Kaabi M, Hussam F, Al-Marsoummi S, Al-Anbaki A, Al-Salihi A, Al-Aubaidy H. Expression of ZO1, vimentin, pan-cadherin and AGTR1 in tanycyte-like cells of the sulcus medianus organum. Biochem Biophys Res Commun 2018; 502:243-249. [PMID: 29803674 DOI: 10.1016/j.bbrc.2018.05.151] [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: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 11/19/2022]
Abstract
Tanycytes are a specialized ependymal lining of brain ventricles with exceptional features of having long basal processes and junctional complexes between cell bodies. These tanycytes are present at the regions of circumventricular organs (CVOs) which possess common morphological and functional features enabling them to be described as the brain windows where the barrier systems have special properties. Previous studies detailed seven of these CVOs but little information is available regarding another putative site at the rostral part of the median sulcus of the 4th ventricle, or the sulcus medianus organum (SMO). Here we performed a pilot immunohistochemical study to support earlier observations suggesting the SMO as a novel CVO. We labeled rat brain with ZO1, vimentin, pan-cadherin and angiotensin II type 1 receptors markers which showed a morphologically distinct population of cells at the region of the SMO similar to tanycytes present in the median eminence, a known CVO. These cells had basal processes reaching the deeply seated blood vessels while the caudal part of the median sulcus did not show similar long cellular extensions. We concluded that tanycyte-like cells are present in the SMO in a pattern resembling that of other CVOs where the strategic location of the SMO is probably for signal integration between brainstem nuclei and the rostrally located neuronal centers.
Collapse
Affiliation(s)
- Muthanna Al-Kaabi
- Al-Nahrain University, College of Medicine, Department of Human Anatomy, Baghdad, Iraq; University of Tasmania, Faculty of Health, School of Medicine, Medical Science Precinct, Hobart, Tasmania, Australia
| | - Fadhil Hussam
- Al-Nahrain University, College of Medicine, Department of Human Anatomy, Baghdad, Iraq
| | - Sarmad Al-Marsoummi
- Al-Nahrain University, College of Medicine, Department of Human Anatomy, Baghdad, Iraq; University of North Dakota, School of Medicine and Health Sciences, Department of Biomedical Sciences, North Dakota, USA
| | - Ali Al-Anbaki
- University of Manchester, Faculty of Biology, Medicine and Health, Manchester, UK
| | - Anam Al-Salihi
- Al-Nahrain University, College of Medicine, Department of Human Anatomy, Baghdad, Iraq
| | - Hayder Al-Aubaidy
- La Trobe University, School of Life Sciences, Department of Physiology, Anatomy & Microbiology, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
98
|
Wang YF, Parpura V. Astroglial Modulation of Hydromineral Balance and Cerebral Edema. Front Mol Neurosci 2018; 11:204. [PMID: 29946238 PMCID: PMC6007284 DOI: 10.3389/fnmol.2018.00204] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Maintenance of hydromineral balance (HB) is an essential condition for life activity at cellular, tissue, organ and system levels. This activity has been considered as a function of the osmotic regulatory system that focuses on hypothalamic vasopressin (VP) neurons, which can reflexively release VP into the brain and blood to meet the demand of HB. Recently, astrocytes have emerged as an essential component of the osmotic regulatory system in addition to functioning as a regulator of the HB at cellular and tissue levels. Astrocytes express all the components of osmoreceptors, including aquaporins, molecules of the extracellular matrix, integrins and transient receptor potential channels, with an operational dynamic range allowing them to detect and respond to osmotic changes, perhaps more efficiently than neurons. The resultant responses, i.e., astroglial morphological and functional plasticity in the supraoptic and paraventricular nuclei, can be conveyed, physically and chemically, to adjacent VP neurons, thereby influencing HB at the system level. In addition, astrocytes, particularly those in the circumventricular organs, are involved not only in VP-mediated osmotic regulation, but also in regulation of other osmolality-modulating hormones, including natriuretic peptides and angiotensin. Thus, astrocytes play a role in local/brain and systemic HB. The adaptive astrocytic reactions to osmotic challenges are associated with signaling events related to the expression of glial fibrillary acidic protein and aquaporin 4 to promote cell survival and repair. However, prolonged osmotic stress can initiate inflammatory and apoptotic signaling processes, leading to glial dysfunction and a variety of brain diseases. Among many diseases of brain injury and hydromineral disorders, cytotoxic and osmotic cerebral edemas are the most common pathological manifestation. Hyponatremia is the most common cause of osmotic cerebral edema. Overly fast correction of hyponatremia could lead to central pontine myelinolysis. Ischemic stroke exemplifies cytotoxic cerebral edema. In this review, we summarize and analyze the osmosensory functions of astrocytes and their implications in cerebral edema.
Collapse
Affiliation(s)
- Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
99
|
Hiratsuka D, Furube E, Taguchi K, Tanaka M, Morita M, Miyata S. Remyelination in the medulla oblongata of adult mouse brain during experimental autoimmune encephalomyelitis. J Neuroimmunol 2018; 319:41-54. [DOI: 10.1016/j.jneuroim.2018.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 10/17/2022]
|
100
|
Erickson MA, Banks WA. Neuroimmune Axes of the Blood-Brain Barriers and Blood-Brain Interfaces: Bases for Physiological Regulation, Disease States, and Pharmacological Interventions. Pharmacol Rev 2018; 70:278-314. [PMID: 29496890 PMCID: PMC5833009 DOI: 10.1124/pr.117.014647] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Central nervous system (CNS) barriers predominantly mediate the immune-privileged status of the brain, and are also important regulators of neuroimmune communication. It is increasingly appreciated that communication between the brain and immune system contributes to physiologic processes, adaptive responses, and disease states. In this review, we discuss the highly specialized features of brain barriers that regulate neuroimmune communication in health and disease. In section I, we discuss the concept of immune privilege, provide working definitions of brain barriers, and outline the historical work that contributed to the understanding of CNS barrier functions. In section II, we discuss the unique anatomic, cellular, and molecular characteristics of the vascular blood-brain barrier (BBB), blood-cerebrospinal fluid barrier, and tanycytic barriers that confer their functions as neuroimmune interfaces. In section III, we consider BBB-mediated neuroimmune functions and interactions categorized as five neuroimmune axes: disruption, responses to immune stimuli, uptake and transport of immunoactive substances, immune cell trafficking, and secretions of immunoactive substances. In section IV, we discuss neuroimmune functions of CNS barriers in physiologic and disease states, as well as pharmacological interventions for CNS diseases. Throughout this review, we highlight many recent advances that have contributed to the modern understanding of CNS barriers and their interface functions.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - William A Banks
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|