Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 126
1.
J Neurosci ; 44(17)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38438259

Oxytocinergic transmission blocks nociception at the peripheral, spinal, and supraspinal levels through the oxytocin receptor (OTR). Indeed, a neuronal pathway from the hypothalamic paraventricular nucleus (PVN) to the spinal cord and trigeminal nucleus caudalis (Sp5c) has been described. Hence, although the trigeminocervical complex (TCC), an anatomical area spanning the Sp5c, C1, and C2 regions, plays a role in some pain disorders associated with craniofacial structures (e.g., migraine), the role of oxytocinergic transmission in modulating nociception at this level has been poorly explored. Hence, in vivo electrophysiological recordings of TCC wide dynamic range (WDR) cells sensitive to stimulation of the periorbital or meningeal region were performed in male Wistar rats. PVN electrical stimulation diminished the neuronal firing evoked by periorbital or meningeal electrical stimulation; this inhibition was reversed by OTR antagonists administered locally. Accordingly, neuronal projections (using Fluoro-Ruby) from the PVN to the WDR cells filled with Neurobiotin were observed. Moreover, colocalization between OTR and calcitonin gene-related peptide (CGRP) or OTR and GABA was found near Neurobiotin-filled WDR cells. Retrograde neuronal tracers deposited at the meningeal (True-Blue, TB) and infraorbital nerves (Fluoro-Gold, FG) showed that at the trigeminal ganglion (TG), some cells were immunopositive to both fluorophores, suggesting that some TG cells send projections via the V1 and V2 trigeminal branches. Together, these data may imply that endogenous oxytocinergic transmission inhibits the nociceptive activity of second-order neurons via OTR activation in CGRPergic (primary afferent fibers) and GABAergic cells.


Electric Stimulation , Oxytocin , Paraventricular Hypothalamic Nucleus , Rats, Wistar , Receptors, Oxytocin , Synaptic Transmission , Animals , Male , Paraventricular Hypothalamic Nucleus/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Oxytocin/metabolism , Oxytocin/analogs & derivatives , Rats , Receptors, Oxytocin/metabolism , Receptors, Oxytocin/antagonists & inhibitors , Synaptic Transmission/physiology , Nociceptors/physiology , Nociceptors/metabolism , Nociception/physiology , Action Potentials/physiology , Action Potentials/drug effects , Meninges/physiology , Neural Inhibition/physiology
2.
Cell Mol Life Sci ; 81(1): 131, 2024 Mar 13.
Article En | MEDLINE | ID: mdl-38472405

The discoveries that cerebrospinal fluid participates in metabolic perivascular exchange with the brain and further drains solutes to meningeal lymphatic vessels have sparked a tremendous interest in translating these seminal findings from animals to humans. A potential two-way coupling between the brain extra-vascular compartment and the peripheral immune system has implications that exceed those concerning neurodegenerative diseases, but also imply that the central nervous system has pushed its immunological borders toward the periphery, where cross-talk mediated by cerebrospinal fluid may play a role in a range of neoplastic and immunological diseases. Due to its non-invasive approach, magnetic resonance imaging has typically been the preferred methodology in attempts to image the glymphatic system and meningeal lymphatics in humans. Even if flourishing, the research field is still in its cradle, and interpretations of imaging findings that topographically associate with reports from animals have yet seemed to downplay the presence of previously described anatomical constituents, particularly in the dura. In this brief review, we illuminate these challenges and assess the evidence for a glymphatic-lymphatic coupling. Finally, we provide a new perspective on how human brain and meningeal clearance function may possibly be measured in future.


Lymphatic Vessels , Animals , Humans , Lymphatic Vessels/metabolism , Central Nervous System , Brain/physiology , Meninges/physiology , Magnetic Resonance Imaging
3.
Cell Mol Life Sci ; 80(11): 332, 2023 Oct 23.
Article En | MEDLINE | ID: mdl-37872442

Meningeal lymphatic vessels (MLVs) help maintain central nervous system (CNS) homeostasis via their ability to facilitate macromolecule waste clearance and neuroimmune trafficking. Although these vessels were overlooked for centuries, they have now been characterized in humans, non-human primates, and rodents. Recent studies in mice have explored the stereotyped growth and expansion of MLVs in dura mater, the various transcriptional, signaling, and environmental factors regulating their development and long-term maintenance, and the pathological changes these vessels undergo in injury, disease, or with aging. Key insights gained from these studies have also been leveraged to develop therapeutic approaches that help augment or restore MLV functions to improve brain health and cognition. Here, we review fundamental processes that control the development of peripheral lymphatic networks and how these might apply to the growth and expansion of MLVs in their unique meningeal environment. We also emphasize key findings in injury and disease models that may reveal additional insights into the plasticity of these vessels throughout the lifespan. Finally, we highlight unanswered questions and future areas of study that can further reveal the exciting therapeutic potential of meningeal lymphatics.


Lymphatic Vessels , Mice , Animals , Lymphatic Vessels/pathology , Meninges/physiology , Central Nervous System , Lymphatic System , Models, Animal
4.
Mol Neurodegener ; 18(1): 55, 2023 08 14.
Article En | MEDLINE | ID: mdl-37580702

Alzheimer's disease (AD) is an aging-related form of dementia associated with the accumulation of pathological aggregates of amyloid beta and neurofibrillary tangles in the brain. These phenomena are accompanied by exacerbated inflammation and marked neuronal loss, which altogether contribute to accelerated cognitive decline. The multifactorial nature of AD, allied to our still limited knowledge of its etiology and pathophysiology, have lessened our capacity to develop effective treatments for AD patients. Over the last few decades, genome wide association studies and biomarker development, alongside mechanistic experiments involving animal models, have identified different immune components that play key roles in the modulation of brain pathology in AD, affecting its progression and severity. As we will relay in this review, much of the recent efforts have been directed to better understanding the role of brain innate immunity, and particularly of microglia. However, and despite the lack of diversity within brain resident immune cells, the brain border tissues, especially the meninges, harbour a considerable number of different types and subtypes of adaptive and innate immune cells. Alongside microglia, which have taken the centre stage as important players in AD research, there is new and exciting evidence pointing to adaptive immune cells, namely T and B cells found in the brain and its meninges, as important modulators of neuroinflammation and neuronal (dys)function in AD. Importantly, a genuine and functional lymphatic vascular network is present around the brain in the outermost meningeal layer, the dura. The meningeal lymphatics are directly connected to the peripheral lymphatic system in different mammalian species, including humans, and play a crucial role in preserving a "healthy" immune surveillance of the CNS, by shaping immune responses, not only locally at the meninges, but also at the level of the brain tissue. In this review, we will provide a comprehensive view on our current knowledge about the meningeal lymphatic vasculature, emphasizing its described roles in modulating CNS fluid and macromolecule drainage, meningeal and brain immunity, as well as glial and neuronal function in aging and in AD.


Alzheimer Disease , Animals , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Genome-Wide Association Study , Meninges/pathology , Meninges/physiology , Lymphatic System/metabolism , Lymphatic System/pathology , Brain/metabolism , Mammals/metabolism
5.
J Pineal Res ; 74(3): e12854, 2023 Apr.
Article En | MEDLINE | ID: mdl-36692235

Photoreceptors in the vertebrate eye are dependent on the retinal pigmented epithelium for a variety of functions including retinal re-isomerization and waste disposal. The light-sensitive pineal gland of fish, birds, and amphibians is evolutionarily related to the eye but lacks a pigmented epithelium. Thus, it is unclear how these functions are performed. Here, we ask whether a subpopulation of zebrafish pineal cells, which express glial markers and visual cycle genes, is involved in maintaining photoreceptors. Selective ablation of these cells leads to a loss of pineal photoreceptors. Moreover, these cells internalize exorhodopsin that is secreted by pineal rod-like photoreceptors, and in turn release CD63-positive extracellular vesicles (EVs) that are taken up by pdgfrb-positive phagocytic cells in the forebrain meninges. These results identify a subpopulation of glial cells that is critical for pineal photoreceptor survival and indicate the existence of cells in the forebrain meninges that receive EVs released by these pineal cells and potentially function in waste disposal.


Neuroglia , Photoreceptor Cells, Vertebrate , Pineal Gland , Visual Perception , Animals , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Gene Expression , Melatonin , Meninges/cytology , Meninges/physiology , Neuroglia/cytology , Neuroglia/metabolism , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/physiology , Pineal Gland/cytology , Pineal Gland/metabolism , Rhodopsin/metabolism , Tetraspanin 30/metabolism , Visual Perception/genetics , Visual Perception/physiology , Zebrafish/genetics , Zebrafish/metabolism
6.
Biomolecules ; 12(6)2022 05 25.
Article En | MEDLINE | ID: mdl-35740873

There is a growing prevalence of vascular cognitive impairment (VCI) worldwide, and most research has suggested that cerebral small vessel disease (CSVD) is the main contributor to VCI. Several potential physiopathologic mechanisms have been proven to be involved in the process of CSVD, such as blood-brain barrier damage, small vessels stiffening, venous collagenosis, cerebral blood flow reduction, white matter rarefaction, chronic ischaemia, neuroinflammation, myelin damage, and subsequent neurodegeneration. However, there still is a limited overall understanding of the sequence and the relative importance of these mechanisms. The glymphatic system (GS) and meningeal lymphatic vessels (mLVs) are the analogs of the lymphatic system in the central nervous system (CNS). As such, these systems play critical roles in regulating cerebrospinal fluid (CSF) and interstitial fluid (ISF) transport, waste clearance, and, potentially, neuroinflammation. Accumulating evidence has suggested that the glymphatic and meningeal lymphatic vessels played vital roles in animal models of CSVD and patients with CSVD. Given the complexity of CSVD, it was significant to understand the underlying interaction between glymphatic and meningeal lymphatic transport with CSVD. Here, we provide a novel framework based on new advances in main four aspects, including vascular risk factors, potential mechanisms, clinical subtypes, and cognition, which aims to explain how the glymphatic system and meningeal lymphatic vessels contribute to the progression of CSVD and proposes a comprehensive insight into the novel therapeutic strategy of CSVD.


Cerebral Small Vessel Diseases , Glymphatic System , Animals , Brain/physiology , Central Nervous System , Glymphatic System/physiology , Humans , Lymphatic System/physiology , Meninges/physiology
7.
Nat Neurosci ; 25(7): 887-899, 2022 07.
Article En | MEDLINE | ID: mdl-35773544

The meninges, comprising the leptomeninges (pia and arachnoid layers) and the pachymeninx (dura layer), participate in central nervous system (CNS) autoimmunity, but their relative contributions remain unclear. Here we report on findings in animal models of CNS autoimmunity and in patients with multiple sclerosis, where, in acute and chronic disease, the leptomeninges were highly inflamed and showed structural changes, while the dura mater was only marginally affected. Although dural vessels were leakier than leptomeningeal vessels, effector T cells adhered more weakly to the dural endothelium. Furthermore, local antigen-presenting cells presented myelin and neuronal autoantigens less efficiently, and the activation of autoreactive T cells was lower in dural than leptomeningeal layers, preventing local inflammatory processes. Direct antigen application was required to evoke a local inflammatory response in the dura. Together, our data demonstrate an uneven involvement of the meningeal layers in CNS autoimmunity, in which effector T cell trafficking and activation are functionally confined to the leptomeninges, while the dura remains largely excluded from CNS autoimmune processes.


Autoimmunity , Meninges , Multiple Sclerosis , Animals , Arachnoid , Central Nervous System , Dura Mater , Humans , Meninges/physiology
8.
Curr Opin Immunol ; 76: 102188, 2022 06.
Article En | MEDLINE | ID: mdl-35569417

The meninges encase the brain and spinal cord and house a variety of immune cells, including developing and mature B cells, and antibody-secreting plasma cells. In homeostasis, these cells localize around the dural venous sinuses, providing a defense 'zone' to protect the brain and spinal cord from blood-borne pathogens. Dural plasma cells predominantly secrete IgA antibodies, and some originate from the gastrointestinal tract, with the number and antibody isotype shaped by the gut microbiome. For developing B cells arriving from the adjacent bone marrow, the dura provides a site to tolerize against central nervous system antigens. In this review, we will discuss our current understanding of meningeal humoral immunity in homeostasis.


Immunity, Humoral , Meninges , Brain , Dura Mater/physiology , Homeostasis , Humans , Meninges/physiology
9.
Curr Opin Hematol ; 29(3): 151-155, 2022 05 01.
Article En | MEDLINE | ID: mdl-35441600

PURPOSE OF REVIEW: The recent (re)discovery of the meningeal lymphatic has brought a new player in brain neurophysiology. This review highlights the state of the current research on the meningeal lymphatic vasculature, from its specific physiology to its increasing implication in normal and pathological brain function. RECENT FINDINGS: Growing evidence are emerging about the uniqueness of the meningeal lymphatic vasculature and its implication in multiple neurological and neurotraumatic disorders. SUMMARY: These studies are highlighting a new and unexpected role for the lymphatic vasculature in brain function and a potential new therapeutic target for neurological disorders.


Lymphatic Vessels , Meninges , Brain , Humans , Lymphatic System , Lymphatic Vessels/physiology , Meninges/pathology , Meninges/physiology
10.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Article En | MEDLINE | ID: mdl-34274976

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Brain/physiology , Ear, Inner/physiology , Heart/physiology , Meninges/physiology , Nervous System/physiopathology , Schwann Cells/physiology , Amphibians/metabolism , Amphibians/physiology , Animals , Brain/metabolism , Cell Lineage/physiology , Ear, Inner/metabolism , Embryonic Development/physiology , Female , Fishes/metabolism , Fishes/physiology , Melanocytes/metabolism , Melanocytes/physiology , Meninges/metabolism , Mice , Nervous System/metabolism , Pregnancy , Receptor, Endothelin B/metabolism , Schwann Cells/metabolism
11.
Science ; 373(6553)2021 07 23.
Article En | MEDLINE | ID: mdl-34083447

The meninges are a membranous structure enveloping the central nervous system (CNS) that host a rich repertoire of immune cells mediating CNS immune surveillance. Here, we report that the mouse meninges contain a pool of monocytes and neutrophils supplied not from the blood but by adjacent skull and vertebral bone marrow. Under pathological conditions, including spinal cord injury and neuroinflammation, CNS-infiltrating myeloid cells can originate from brain borders and display transcriptional signatures distinct from their blood-derived counterparts. Thus, CNS borders are populated by myeloid cells from adjacent bone marrow niches, strategically placed to supply innate immune cells under homeostatic and pathological conditions. These findings call for a reinterpretation of immune-cell infiltration into the CNS during injury and autoimmunity and may inform future therapeutic approaches that harness meningeal immune cells.


Bone Marrow Cells/physiology , Central Nervous System Diseases/immunology , Central Nervous System/immunology , Meninges/immunology , Myeloid Cells/physiology , Skull/anatomy & histology , Spine/anatomy & histology , Animals , Bone Marrow/physiology , Brain/cytology , Brain/immunology , Brain/physiology , Cell Movement , Central Nervous System/cytology , Central Nervous System Diseases/pathology , Dura Mater/cytology , Dura Mater/immunology , Dura Mater/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Homeostasis , Meninges/cytology , Meninges/physiology , Mice , Monocytes/physiology , Neutrophils/physiology , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord/physiology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
12.
J Cereb Blood Flow Metab ; 41(8): 1842-1857, 2021 08.
Article En | MEDLINE | ID: mdl-33444089

The distribution and clearance of erythrocytes after subarachnoid hemorrhage (SAH) is poorly understood. We aimed to characterize the distribution of erythrocytes after SAH and the cells involved in their clearance. To visualize erythrocyte distribution, we injected fluorescently-labelled erythrocytes into the prechiasmatic cistern of mice. 10 minutes after injection, we found labelled erythrocytes in the subarachnoid space and ventricular system, and also in the perivascular spaces surrounding large penetrating arterioles. 2 and 5 days after SAH, fluorescence was confined within leptomeningeal and perivascular cells. We identified the perivascular cells as perivascular macrophages based on their morphology, location, Iba-1 immunoreactivity and preferential uptake of FITC-dextran. We subsequently depleted meningeal and perivascular macrophages 2 days before or 3 hours after SAH with clodronate liposomes. At day 5 after SAH, we found increased blood deposition in mice treated prior to SAH, but not those treated after. Treatment post-SAH improved neurological scoring, reduced neuronal cell death and perivascular inflammation, whereas pre-treatment only reduced perivascular inflammation. Our data indicate that after SAH, erythrocytes are distributed throughout the subarachnoid space extending into the perivascular spaces of parenchymal arterioles. Furthermore, meningeal and perivascular macrophages are involved in erythrocyte uptake and play an important role in outcome after SAH.


Macrophages/physiology , Subarachnoid Hemorrhage/pathology , Animals , Brain/pathology , Disease Models, Animal , Erythrocytes/chemistry , Erythrocytes/cytology , Erythrocytes/metabolism , Gliosis , Glymphatic System/cytology , Glymphatic System/pathology , Macrophages/cytology , Male , Meninges/cytology , Meninges/physiology , Mice , Neurons/metabolism , Neurons/pathology , Optical Imaging , Subarachnoid Hemorrhage/metabolism , Subarachnoid Space/cytology , Subarachnoid Space/pathology
13.
Stem Cells ; 39(2): 227-239, 2021 02.
Article En | MEDLINE | ID: mdl-33270951

Although the bone marrow contains most hematopoietic activity during adulthood, hematopoietic stem and progenitor cells can be recovered from various extramedullary sites. Cells with hematopoietic progenitor properties have even been reported in the adult brain under steady-state conditions, but their nature and localization remain insufficiently defined. Here, we describe a heterogeneous population of myeloid progenitors in the leptomeninges of adult C57BL/6 mice. This cell pool included common myeloid, granulocyte/macrophage, and megakaryocyte/erythrocyte progenitors. Accordingly, it gave rise to all major myelo-erythroid lineages in clonogenic culture assays. Brain-associated progenitors persisted after tissue perfusion and were partially inaccessible to intravenous antibodies, suggesting their localization behind continuous blood vessel endothelium such as the blood-arachnoid barrier. Flt3Cre lineage tracing and bone marrow transplantation showed that the precursors were derived from adult hematopoietic stem cells and were most likely continuously replaced via cell trafficking. Importantly, their occurrence was tied to the immunologic state of the central nervous system (CNS) and was diminished in the context of neuroinflammation and ischemic stroke. Our findings confirm the presence of myeloid progenitors at the meningeal border of the brain and lay the foundation to unravel their possible functions in CNS surveillance and local immune cell production.


Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Brain/physiology , Cell Differentiation/physiology , Meninges/physiology , Meninges/transplantation , Age Factors , Animals , Bone Marrow/physiology , Brain/cytology , Female , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Male , Meninges/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Circ Res ; 128(1): 42-58, 2021 01 08.
Article En | MEDLINE | ID: mdl-33135960

RATIONALE: The recent discovery of meningeal lymphatics in mammals is reshaping our understanding of fluid homeostasis and cellular waste management in the brain, but visualization and experimental analysis of these vessels is challenging in mammals. Although the optical clarity and experimental advantages of zebrafish have made this an essential model organism for studying lymphatic development, the existence of meningeal lymphatics has not yet been reported in this species. OBJECTIVE: Examine the intracranial space of larval, juvenile, and adult zebrafish to determine whether and where intracranial lymphatic vessels are present. METHODS AND RESULTS: Using high-resolution optical imaging of the meninges in living animals, we show that zebrafish possess a meningeal lymphatic network comparable to that found in mammals. We confirm that this network is separate from the blood vascular network and that it drains interstitial fluid from the brain. We document the developmental origins and growth of these vessels into a distinct network separated from the external lymphatics. Finally, we show that these vessels contain immune cells and perform live imaging of immune cell trafficking and transmigration in meningeal lymphatics. CONCLUSIONS: This discovery establishes the zebrafish as a important new model for experimental analysis of meningeal lymphatic development and opens up new avenues for probing meningeal lymphatic function in health and disease.


Lymphangiogenesis , Lymphatic Vessels/physiology , Meninges/physiology , Microscopy, Confocal , Optical Imaging , Animals , Animals, Genetically Modified , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Lymphatic Vessels/immunology , Meninges/immunology , Neutrophil Infiltration , Neutrophils/immunology , Vascular Endothelial Growth Factor C/pharmacology , Zebrafish/genetics
15.
J Neurotrauma ; 38(13): 1748-1761, 2021 06 01.
Article En | MEDLINE | ID: mdl-33191848

The meninges are membranous tissues that are pivotal in maintaining homeostasis of the central nervous system. Despite the importance of the cranial meninges in nervous system physiology and in head injury mechanics, our knowledge of the tissues' mechanical behavior and structural composition is limited. This systematic review analyzes the existing literature on the mechanical properties of the meningeal tissues. Publications were identified from a search of Scopus, Academic Search Complete, and Web of Science and screened for eligibility according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The review details the wide range of testing techniques employed to date and the significant variability in the observed experimental findings. Our findings identify many gaps in the current literature that can serve as a guide for future work for meningeal mechanics investigators. The review identifies no peer-reviewed mechanical data on the falx and tentorium tissues, both of which have been identified as key structures in influencing brain injury mechanics. A dearth of mechanical data for the pia-arachnoid complex also was identified (no experimental mechanics studies on the human pia-arachnoid complex were identified), which is desirable for biofidelic modeling of human head injuries. Finally, this review provides recommendations on how experiments can be conducted to allow for standardization of test methodologies, enabling simplified comparisons and conclusions on meningeal mechanics.


Arachnoid/physiology , Biomechanical Phenomena/physiology , Dura Mater/physiology , Pia Mater/physiology , Animals , Arachnoid/cytology , Brain/cytology , Brain/physiology , Dura Mater/cytology , Humans , Meninges/cytology , Meninges/physiology , Pia Mater/cytology
16.
Annu Rev Immunol ; 38: 597-620, 2020 04 26.
Article En | MEDLINE | ID: mdl-32340575

Neuroimmunology, albeit a relatively established discipline, has recently sparked numerous exciting findings on microglia, the resident macrophages of the central nervous system (CNS). This review addresses meningeal immunity, a less-studied aspect of neuroimmune interactions. The meninges, a triple layer of membranes-the pia mater, arachnoid mater, and dura mater-surround the CNS, encompassing the cerebrospinal fluid produced by the choroid plexus epithelium. Unlike the adjacent brain parenchyma, the meninges contain a wide repertoire of immune cells. These constitute meningeal immunity, which is primarily concerned with immune surveillance of the CNS, and-according to recent evidence-also participates in postinjury CNS recovery, chronic neurodegenerative conditions, and even higher brain function. Meningeal immunity has recently come under the spotlight owing to the characterization of meningeal lymphatic vessels draining the CNS. Here, we review the current state of our understanding of meningeal immunity and its effects on healthy and diseased brains.


Central Nervous System/immunology , Central Nervous System/metabolism , Disease Susceptibility , Homeostasis , Immunity , Meninges/physiology , Animals , Humans , Lymphatic Vessels/immunology , Lymphatic Vessels/metabolism , Neuroimmunomodulation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
17.
Biomech Model Mechanobiol ; 19(3): 875-892, 2020 Jun.
Article En | MEDLINE | ID: mdl-31745681

Blast-induced traumatic brain injury (bTBI) is a critical health concern. This issue is being addressed in terms of identifying a cause-effect relationship between the mechanical insult in the form of a blast and resulting injury to the brain. Understanding wave propagation through the head is an important aspect in this regard. The objective of this work was to study the blast wave propagation through the layered architecture of the head with an emphasis on understanding the wave transmission mechanism. Toward this end, one-dimensional (1D) finite element head model is built for a simplified surrogate, human, and rat. Motivated from experimental investigations, four different head layer configurations have been considered. These configurations are: (A) Skull-Brain, (B) Skin-Skull-Brain, (C) Skin-Skull-Dura-Arachnoid-CSF-Pia-Brain, (D) Skin-Skull-Dura-Arachnoid-AT-Pia-Brain. The validated head model is subjected to flattop and Friedlander loading implied in the blast, and the resulting response is evaluated in terms of brain pressures. Our results suggest that wave propagation through head parenchyma plays an important role in blast wave transmission. The thickness, material properties of head layers, and rise time of an input pulse govern the temporal evolution of pressure in the brain. The key findings of this work are: (a) Skin and meninges amplify the applied input pressure, whereas air sinus has an attenuation effect. (b) Model is able to describe experimentally recorded peak pressures and rise times in the brain, including variations within the aforementioned experimental head models of TBI. This reinforces that the wave transmission is an important loading pathway to the brain. (c) Equivalent layer theory for modeling meningeal layers as a single layer has been proposed, and it gives reasonable agreement with each meningeal layer modeled explicitly. This modeling approach has a great utility in 3D head models. The potential applications of 1D head model in evaluation of new helmet materials, brain sensor calibration, and brain pressure estimation for a given explosive strength have also been demonstrated. Overall, these results provide important insights into the understanding of mechanics of blast wave transmission in the head.


Blast Injuries/physiopathology , Brain Injuries/physiopathology , Brain/physiopathology , Animals , Arachnoid/physiology , Biomechanical Phenomena , Brain/physiology , Computer Simulation , Dura Mater/physiology , Elasticity , Finite Element Analysis , Head/physiology , Humans , Intracranial Pressure , Meninges/physiology , Models, Biological , Pia Mater/physiology , Rats , Skull/physiology
18.
Neurosurg Rev ; 43(4): 1055-1064, 2020 Aug.
Article En | MEDLINE | ID: mdl-31209659

Lymphangiogenesis is associated with some pathological conditions such as inflammation, tissue repair, and tumor growth. Recently, a paradigm shift occurred following the discovery of meningeal lymphatic structures in the human central nervous system (CNS); these structures may be a key drainage route for cerebrospinal fluid (CSF) into the peripheral blood and may also contribute to inflammatory reaction and immune surveillance of the CNS. Lymphatic vessels located along the dural sinuses absorb CSF from the adjacent subarachnoid space and brain interstitial fluid via the glymphatic system, which is composed of aquaporin-4 water channels expressed on perivascular astrocytic end-feet membranes. Magnetic resonance imaging (MRI) clearly visualized these lymphatic vessels in the human dura mater. The conception of some neurological disorders, such as multiple sclerosis and Alzheimer's disease, has been changed by this paradigm shift. Meningeal lymphatic vessels could be a promising therapeutic target for the prevention of neurological disorders. However, the involvement of meningeal lymphatic vessels in the pathophysiology has not been fully elucidated and is the subject of future investigations. In this article, to understand the involvement of meningeal lymphatic vessels in neurological disorders, we review the differences between lymphangiogenesis in the CNS and in other tissues during both developmental and adulthood stages, and pathological conditions that may be associated with meningeal lymphatic vessels in the CNS.


Central Nervous System/physiology , Lymphatic Vessels/physiology , Central Nervous System/diagnostic imaging , Central Nervous System/physiopathology , Glymphatic System , Humans , Lymphatic Vessels/diagnostic imaging , Lymphatic Vessels/physiopathology , Meninges/diagnostic imaging , Meninges/physiology , Meninges/physiopathology
19.
J Neural Eng ; 17(1): 016027, 2020 01 14.
Article En | MEDLINE | ID: mdl-31689695

OBJECTIVE: Understanding how current reaches the brain during transcranial electrical stimulation (tES) underpins efforts to rationalize outcomes and optimize interventions. To this end, computational models of current flow relate applied dose to brain electric field. Conventional tES modeling considers distinct tissues like scalp, skull, cerebrospinal fluid (CSF), gray matter and white matter. The properties of highly conductive CSF are especially important. However, modeling the space between skull and brain as entirely CSF is not an accurate representation of anatomy. The space conventionally modeled as CSF is approximately half meninges (dura, arachnoid, and pia) with lower conductivity. However, the resolution required to describe individual meningeal layers is computationally restrictive in an MRI-derived head model. Emulating the effect of meninges through CSF conductivity modification could improve accuracy with minimal cost. APPROACH: Models with meningeal layers were developed in a concentric sphere head model. Then, in a model with only CSF between skull and brain, CSF conductivity was optimized to emulate the effect of meningeal layers on cortical electric field for multiple electrode positions. This emulated conductivity was applied to MRI-derived models. MAIN RESULTS: Compared to a model with conventional CSF conductivity (1.65 S m-1), emulated CSF conductivity (0.85 S m-1) produced voltage fields better correlated with intracranial recordings from epilepsy patients. SIGNIFICANCE: Conventional tES models have been validated using intracranial recording. Residual errors may nonetheless impact model utility. Because CSF is so conductive to current flow, misrepresentation of the skull-brain interface as entirely CSF is not realistic for tES modeling. Updating the conventional model with a CSF conductivity emulating the effect of the meninges enhances modeling accuracy without increasing model complexity. This allows existing modeling pipelines to be leveraged with a simple conductivity change. Using 0.85 S m-1 emulated CSF conductivity is recommended as the new standard in non-invasive brain stimulation modeling.


Finite Element Analysis , Meninges/anatomy & histology , Meninges/physiology , Models, Neurological , Transcranial Direct Current Stimulation/methods , Humans , Magnetic Resonance Imaging/methods , Meninges/diagnostic imaging , Skull/anatomy & histology , Skull/diagnostic imaging , Skull/physiology
20.
Mater Sci Eng C Mater Biol Appl ; 102: 34-44, 2019 Sep.
Article En | MEDLINE | ID: mdl-31147006

Spinal cord injury (SCI) is one of the most precarious conditions which have been one of the major reasons for continuous increasing mortality rate of SCI patients. Currently, there is no effective treatment modality for SCI patients posing major threat to the scientific and medical community. The available strategies don't mimic with the natural processes of nervous tissues repair/regeneration and majority of the approaches may induce the additional fibrotic or immunological response at the injury site and are not readily available on demand. To overcome these hurdles, we have developed a ready to use bioengineered human functional neurological construct (BHNC) for regenerative applications in SCI defects. We used cryopreserved meningeal tissues (CMT) for bioengineering these neurological constructs using acellularization and repopulation technology. The technology adopted herein generates intact neurological scaffolds from CMT and retains several crucial structural, biochemical and mechanical cues to enhance the regenerative mechanisms. The neurogenic differentiation on CMT scaffolds was almost similar to the freshly prepared meningeal scaffolds and mimics with the natural nervous tissue developmental mechanisms which offer intact 3D-microarchitecture and hospitable microenvironment enriched with several crucial neurotrophins for long-term cell survival and function. Functional assessment of developed BHNC showed highly increased positive staining for pre-synaptic granules of Synapsis-1 along with MAP-2 antibody with punctuate distribution in axonal regions of the neuronal cells which was well supported by the gene expression analysis of functional transcripts. Given the significant improvement in the field may enable to generate more such ready to use functional BHNC for wider applicability in SCI repair/regeneration.


Biomimetic Materials/pharmacology , Cryopreservation , Meninges/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biomechanical Phenomena , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Humans , Meninges/drug effects , Meninges/ultrastructure , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism
...