Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
Add more filters

Publication year range
1.
J Neuroinflammation ; 21(1): 165, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937750

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a significant risk factor for Alzheimer's disease (AD), and accumulating evidence supports a role for adaptive immune B and T cells in both TBI and AD pathogenesis. We previously identified B cell and major histocompatibility complex class II (MHCII)-associated invariant chain peptide (CLIP)-positive B cell expansion after TBI. We also showed that antagonizing CLIP binding to the antigen presenting groove of MHCII after TBI acutely reduced CLIP + splenic B cells and was neuroprotective. The current study investigated the chronic effects of antagonizing CLIP in the 5xFAD Alzheimer's mouse model, with and without TBI. METHODS: 12-week-old male wild type (WT) and 5xFAD mice were administered either CLIP antagonist peptide (CAP) or vehicle, once at 30 min after either sham or a lateral fluid percussion injury (FPI). Analyses included flow cytometric analysis of immune cells in dural meninges and spleen, histopathological analysis of the brain, magnetic resonance diffusion tensor imaging, cerebrovascular analysis, and assessment of motor and neurobehavioral function over the ensuing 6 months. RESULTS: 9-month-old 5xFAD mice had significantly more CLIP + B cells in the meninges compared to age-matched WT mice. A one-time treatment with CAP significantly reduced this population in 5xFAD mice. Importantly, CAP also improved some of the immune, histopathological, and neurobehavioral impairments in 5xFAD mice over the ensuing six months. Although FPI did not further elevate meningeal CLIP + B cells, it did negate the ability of CAP to reduce meningeal CLIP + B cells in the 5xFAD mice. FPI at 3 months of age exacerbated some aspects of AD pathology in 5xFAD mice, including further reducing hippocampal neurogenesis, increasing plaque deposition in CA3, altering microgliosis, and disrupting the cerebrovascular structure. CAP treatment after injury ameliorated some but not all of these FPI effects.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , B-Lymphocytes , Brain Injuries, Traumatic , Histocompatibility Antigens Class II , Mice, Transgenic , Animals , Mice , Male , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/drug therapy , Histocompatibility Antigens Class II/metabolism , B-Lymphocytes/drug effects , Meninges/pathology , Meninges/drug effects , Amyloid beta-Protein Precursor/genetics , Alzheimer Disease/pathology , Alzheimer Disease/drug therapy , Humans , Disease Models, Animal , Presenilin-1/genetics , Mice, Inbred C57BL
2.
Cephalalgia ; 44(9): 3331024241281493, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39233656

ABSTRACT

BACKGROUND: We wished to explore possible sexual dimorphism in mechanisms sensitizing or activating meningeal nociceptors that can promote the headache phase of migraine. METHODS: Male and female C57BL6J mice received either supradural orexin B and an inflammatory mediator cocktail (IM) with migraine-like pain behaviors and photophobia recorded. Expression of orexin 2 receptor (OX2R) in trigeminal ganglion (TG) and phosphorylated extracellular signal-regulated kinases (ERK) levels in trigeminal nucleus caudalis (TNC) were evaluated. Orexin B-induced excitability of TG cells was assessed with patch-clamp electrophysiology. Intranasal delivery of CRISPR/Cas9 plasmids was used to edit the expression of OX2R in the TG. RESULTS: Supradural orexin B induced migraine-like pain behaviors, photophobia and increased TNC ERK phosphorylation exclusively in males. Blockade of orexin signaling with supradural suvorexant, a dual orexin receptor antagonist, prevented, but did not reverse, migraine-like pain in males induced by supradural IM cocktail. OX2R expression was higher in male TG and orexin B increased TG neuron excitability in males. Intranasal OX2R CRISPR/Cas9 reduced TG receptor expression and orexin B-induced TNC ERK phosphorylation and prevented migraine-like pain induced by supradural orexin B in males. CONCLUSIONS: Our studies reveal a male-specific mechanism of TG nociceptor sensitization and migraine-like pain behavior mediated by orexin B/OX2R signaling. Sexually dimorphic mechanisms of trigeminal nociceptor sensitization and activation offer opportunities to improve patient outcomes by considering patient sex and may influence clinical trial design and interpretation.


Subject(s)
Mice, Inbred C57BL , Migraine Disorders , Orexin Receptors , Trigeminal Ganglion , Animals , Male , Female , Mice , Migraine Disorders/metabolism , Migraine Disorders/physiopathology , Orexin Receptors/metabolism , Orexin Receptors/genetics , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/drug effects , Meninges/drug effects , Meninges/metabolism , Sex Characteristics , Orexins/metabolism
3.
Stroke ; 53(3): 987-998, 2022 03.
Article in English | MEDLINE | ID: mdl-35144488

ABSTRACT

BACKGROUND: Promotion of hematoma resolution in a timely manner reduces intracerebral hemorrhage (ICH) brain injury induced by toxic blood components and subsequent neuroinflammation. The meningeal lymphatic system is responsible for clearance of macromolecules and pathogenic substances from the central nervous system; however, its role in intraparenchymal hematoma clearance and ICH outcomes is unknown. In the present study, we aimed to understand the contribution of the meningeal lymphatic system to ICH pathologies and to test whether pharmacological enhancement of meningeal lymphatic function promotes hematoma resolution and brain recovery after ICH. METHODS: Immunofluorescence of whole-mount meninges was used to measure complexity and coverage level of meningeal lymphatic vasculature following ICH induction. Fluorescent microbeads and PKH-26-labeled erythrocytes were used to evaluate drainage function of the meningeal lymphatic system. Visudyne treatment, deep cervical lymph node ligation, and VEGF (vascular endothelial growth factor)-C injection were performed to manipulate meningeal lymphatic function. Neurobehavioral performance and hematoma volume were assayed by the cylinder test and histological measurements. Iron deposition, residual erythrocytes, neuronal loss, and astrogliosis were assessed by immunohistochemistry and antibody-based fluorescence staining. RESULTS: Meningeal lymphangiogenesis and enhanced lymphatic drainage occurred during the late phase of ICH. Ablation and blockage of meningeal lymphatic vessels impeded hematoma clearance, whereas pharmacological enhancement of their function reduced hematoma volume, improved behavioral performance, and reduced brain residual erythrocytes, iron deposition, neuronal loss, and astroglial activation. CONCLUSIONS: Early enhancement of meningeal lymphatic function is beneficial for ICH recovery. Targeting the meningeal lymphatic system is therefore a potential therapeutic approach for treating ICH.


Subject(s)
Brain/pathology , Cerebral Hemorrhage/pathology , Lymphangiogenesis/physiology , Lymphatic System/pathology , Meninges/pathology , Animals , Brain/drug effects , Cerebral Hemorrhage/drug therapy , Cilostazol/pharmacology , Cilostazol/therapeutic use , Lymphangiogenesis/drug effects , Lymphatic System/drug effects , Male , Meninges/drug effects , Mice , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
4.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34638999

ABSTRACT

Neural precursors (NPs) present in the hippocampus can be modulated by several neurogenic stimuli, including environmental enrichment (EE) acting through BDNF-TrkB signaling. We have recently identified NPs in meninges; however, the meningeal niche response to pro-neurogenic stimuli has never been investigated. To this aim, we analyzed the effects of EE exposure on NP distribution in mouse brain meninges. Following neurogenic stimuli, although we did not detect modification of the meningeal cell number and proliferation, we observed an increased number of neural precursors in the meninges. A lineage tracing experiment suggested that EE-induced ß3-Tubulin+ immature neuronal cells present in the meninges originated, at least in part, from GLAST+ radial glia cells. To investigate the molecular mechanism responsible for meningeal reaction to EE exposure, we studied the BDNF-TrkB interaction. Treatment with ANA-12, a TrkB non-competitive inhibitor, abolished the EE-induced meningeal niche changes. Overall, these data showed, for the first time, that EE exposure induced meningeal niche remodeling through TrkB-mediated signaling. Fluoxetine treatment further confirmed the meningeal niche response, suggesting it may also respond to other pharmacological neurogenic stimuli. A better understanding of the neurogenic stimuli modulation for meninges may be useful to improve the effectiveness of neurodegenerative and neuropsychiatric treatments.


Subject(s)
Cellular Microenvironment , Environment , Membrane Glycoproteins/metabolism , Meninges/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Biomarkers , Brain-Derived Neurotrophic Factor/metabolism , Fluorescent Antibody Technique , Fluoxetine/pharmacology , Meninges/drug effects , Meninges/pathology , Mice , Neuroglia/metabolism , Neurons/metabolism
5.
Nature ; 505(7482): 223-8, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24317693

ABSTRACT

Traumatic brain injury (TBI) is increasingly appreciated to be highly prevalent and deleterious to neurological function. At present, no effective treatment options are available, and little is known about the complex cellular response to TBI during its acute phase. To gain insights into TBI pathogenesis, we developed a novel murine closed-skull brain injury model that mirrors some pathological features associated with mild TBI in humans and used long-term intravital microscopy to study the dynamics of the injury response from its inception. Here we demonstrate that acute brain injury induces vascular damage, meningeal cell death, and the generation of reactive oxygen species (ROS) that ultimately breach the glial limitans and promote spread of the injury into the parenchyma. In response, the brain elicits a neuroprotective, purinergic-receptor-dependent inflammatory response characterized by meningeal neutrophil swarming and microglial reconstitution of the damaged glial limitans. We also show that the skull bone is permeable to small-molecular-weight compounds, and use this delivery route to modulate inflammation and therapeutically ameliorate brain injury through transcranial administration of the ROS scavenger, glutathione. Our results shed light on the acute cellular response to TBI and provide a means to locally deliver therapeutic compounds to the site of injury.


Subject(s)
Brain Injuries/complications , Brain Injuries/pathology , Encephalitis/pathology , Encephalitis/prevention & control , Administration, Topical , Animals , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Astrocytes/pathology , Brain/drug effects , Brain/pathology , Brain Injuries/diagnosis , Brain Injuries/drug therapy , Cell Death/drug effects , Disease Models, Animal , Encephalitis/complications , Encephalitis/drug therapy , Glasgow Coma Scale , Glutathione/administration & dosage , Glutathione/therapeutic use , Humans , Intracranial Hemorrhages/complications , Intracranial Hemorrhages/diagnosis , Male , Meninges/drug effects , Meninges/pathology , Mice , Microglia/cytology , Microglia/drug effects , Microglia/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Neutrophils/drug effects , Neutrophils/physiology , Purinergic P2 Receptor Antagonists/administration & dosage , Purinergic P2 Receptor Antagonists/pharmacology , Purinergic P2 Receptor Antagonists/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X7/metabolism , Skull/metabolism
6.
Cephalalgia ; 39(11): 1358-1365, 2019 10.
Article in English | MEDLINE | ID: mdl-31475573

ABSTRACT

BACKGROUND: Botulinum neurotoxin type A, an FDA-approved prophylactic drug for chronic migraine, is thought to achieve its therapeutic effect through blocking activation of unmyelinated meningeal nociceptors and their downstream communications with myelinated nociceptors and potentially the vasculature and immune cells. Prior investigations to determine botulinum neurotoxin type A effects on meningeal nociceptors were carried out in male rats and tested with stimuli that act outside the blood brain barrier. Here, we sought to explore the effects of extracranial injections of botulinum neurotoxin type A on activation of meningeal nociceptors by cortical spreading depression, an event which occurs inside the blood brain barrier, in female rats. MATERIAL AND METHODS: Using single-unit recording, we studied myelinated C- and unmyelinated Aδ-meningeal nociceptors' responses to cortical spreading depression 7-14 days after injection of botulinum neurotoxin type A or saline along calvarial sutures. RESULTS: In female rats, responses to cortical spreading depression were typically more prolonged and, in some cases, began at relatively longer latencies post-cortical spreading depression, than had been observed in previous studies in male rats. Extracranial administration of botulinum neurotoxin type A reduced significantly the prolonged firing of the meningeal nociceptors, in the combined sample of Aδ- and C-fiber, but not their response probability. DISCUSSION: The findings suggest that the mechanism of action by which botulinum neurotoxin type A prevents migraine differ from the one by which calcitonin gene-related peptide monoclonal antibodies prevent migraine and that even when the origin of migraine is central (i.e. in the cortex), a peripherally acting drug can intercept/prevent the headache.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Cortical Spreading Depression/drug effects , Meninges/drug effects , Neuromuscular Agents/pharmacology , Nociceptors/drug effects , Animals , Female , Nerve Fibers, Unmyelinated/drug effects , Rats , Rats, Sprague-Dawley
7.
J Stroke Cerebrovasc Dis ; 27(5): 1237-1251, 2018 May.
Article in English | MEDLINE | ID: mdl-29337049

ABSTRACT

BACKGROUND: Development of collateral circulation after acute ischemic stroke is triggered by shear stress that occurs in pre-existing arterioles. Recently, sphingosine-1-phosphate receptor 1 (S1P1) on endothelial cells was reported to sense shear stress and transduce its signaling pathways. METHODS: BALB/c mice (n = 118) were subjected to permanent middle cerebral artery occlusion (pMCAO) or sham operation. We investigated the effect of an S1P1-selective agonist SEW2871 on leptomeningeal collateral arteries and neurological outcome after pMCAO. RESULTS: Immunohistochemistry showed that without treatment, the expression of S1P1 on endothelial cells of leptomeningeal arteries and capillaries increased early after pMCAO, peaking at 6 hours, whereas a significant increase in the expression of S1P1 in neurons was seen from 24 hours later. After intraperitoneal administration of SEW2871 for 7 days after pMCAO, the number of leptomeningeal collateral arteries was significantly increased, cerebral blood flow improved, infarct volume was decreased, and neurological outcome improved compared with the controls. Significantly increased phosphorylation of endothelial nitric oxide synthase (eNOS) as early as 6 hours after pMCAO and higher expression of tight junction proteins at postoperative day 3 were observed with SEW2871 treatment as assessed by Western blot. Daily administration of SEW2871 also increased capillary density in peri-infarct regions and promoted monocyte/macrophage mobilization to the surface of ischemic cortex at 7 days after pMCAO. CONCLUSIONS: An S1P1-selective agonist enhanced leptomeningeal collateral circulation via eNOS phosphorylation and promoted postischemic angiogenesis with reinforced blood-brain barrier integrity in a mouse model of acute ischemic stroke, leading to smaller infarct volume and better neurological outcome.


Subject(s)
Cerebrovascular Circulation/drug effects , Collateral Circulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Meninges/blood supply , Meninges/drug effects , Neovascularization, Physiologic/drug effects , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Thiophenes/pharmacology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cell Line , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Macrophages/drug effects , Macrophages/metabolism , Male , Meninges/metabolism , Meninges/pathology , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Receptors, Lysosphingolipid/metabolism , Recovery of Function , Signal Transduction/drug effects , Sphingosine-1-Phosphate Receptors , Tight Junction Proteins/metabolism , Time Factors
8.
Dev Biol ; 420(1): 148-165, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27671872

ABSTRACT

Growth and maturation of the cerebrovasculature is a vital event in neocortical development however mechanisms that control cerebrovascular development remain poorly understood. Mutations in or deletions that include the FOXC1 gene are associated with congenital cerebrovascular anomalies and increased stroke risk in patients. Foxc1 mutant mice display severe cerebrovascular hemorrhage at late gestational ages. While these data demonstrate Foxc1 is required for cerebrovascular development, its broad expression in the brain vasculature combined with Foxc1 mutant's complex developmental defects have made it difficult to pinpoint its function(s). Using global and conditional Foxc1 mutants, we find 1) significant cerebrovascular growth defects precede cerebral hemorrhage and 2) expression of Foxc1 in neural crest-derived meninges and brain pericytes, though not endothelial cells, is required for normal cerebrovascular development. We provide evidence that reduced levels of meninges-derived retinoic acid (RA), caused by defects in meninges formation in Foxc1 mutants, is a major contributing factor to the cerebrovascular growth defects in Foxc1 mutants. We provide data that suggests that meninges-derived RA ensures adequate growth of the neocortical vasculature via regulating expression of WNT pathway proteins and neural progenitor derived-VEGF-A. Our findings offer the first evidence for a role of the meninges in brain vascular development and provide new insight into potential causes of cerebrovascular defects in patients with FOXC1 mutations.


Subject(s)
Brain/abnormalities , Forkhead Transcription Factors/genetics , Meninges/metabolism , Mutation/genetics , Signal Transduction , Tretinoin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/metabolism , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Brain/blood supply , Brain/pathology , Cells, Cultured , Cerebral Hemorrhage/pathology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Embryo, Mammalian/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Integrases/metabolism , Meninges/drug effects , Mice , Neocortex/blood supply , Neocortex/embryology , Neocortex/pathology , Pericytes/drug effects , Pericytes/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology , beta-Galactosidase/metabolism
9.
J Neuroinflammation ; 14(1): 32, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183352

ABSTRACT

BACKGROUND: The spatial learning abilities of developing mice benefit from extrinsic cues, such as an enriched environment, with concomitant enhancement in cognitive functions. Interestingly, such enhancements can be further increased through intrinsic Bacillus Calmette-Guérin (BCG) vaccination. RESULTS: Here, we first report that combined neonatal BCG vaccination and exposure to an enriched environment (Enr) induced combined neurobeneficial effects, including hippocampal long-term potentiation, and increased neurogenesis and spatial learning and memory, in mice exposed to the Enr and vaccinated with BCG relative to those in the Enr that did not receive BCG vaccination. Neonatal BCG vaccination markedly induced anti-inflammatory meningeal macrophage polarization both in regular and Enr breeding mice. The meninges are composed of the pia mater, dura mater, and choroid plexus. Alternatively, this anti-inflammatory activity of the meninges occurred simultaneously with increased expression of the neurotrophic factors BDNF/IGF-1 and the M2 microglial phenotype in the hippocampus. Our results reveal a critical role for BCG vaccination in the regulation of neurogenesis and spatial cognition through meningeal macrophage M2 polarization and neurotrophic factor expression; these effects were completely or partially prevented by minocycline or anti-IL-10 antibody treatment, respectively. CONCLUSIONS: Together, we first claim that immunological factor and environmental factor induce a combined effect on neurogenesis and cognition via a common pathway-meningeal macrophage M2 polarization. We also present a novel functional association between peripheral T lymphocytes and meningeal macrophages after evoking adaptive immune responses in the periphery whereby T lymphocytes are recruited to the meninges in response to systemic IFN-γ signaling. This leads to meningeal macrophage M2 polarization, subsequent to microglial M2 activation and neurotrophic factor expression, and eventually promotes a positive behavior.


Subject(s)
BCG Vaccine/administration & dosage , Cognition/physiology , Environment , Macrophages/metabolism , Neurogenesis/physiology , Spatial Behavior/physiology , Animals , Animals, Newborn , Cell Polarity/drug effects , Cell Polarity/physiology , Cognition/drug effects , Female , Macrophages/drug effects , Macrophages/immunology , Male , Maze Learning/drug effects , Maze Learning/physiology , Meninges/drug effects , Meninges/immunology , Meninges/metabolism , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Spatial Behavior/drug effects
10.
Mult Scler ; 22(7): 972-4, 2016 06.
Article in English | MEDLINE | ID: mdl-27207452

ABSTRACT

BACKGROUND: Contrast-enhanced (ce) fluid-attenuated inversion recovery magnetic resonance imaging (FLAIR MRI) has recently been shown to identify leptomeningeal pathology in multiple sclerosis. OBJECTIVE: To demonstrate leptomeningeal enhancement on three-dimensional (3D) FLAIR in a case of Susac's syndrome. METHODS: Leptomeningeal enhancement was correlated with clinical activity over 20 months and compared to retinal fluorescein angiography. RESULTS: The size, number, and location of leptomeningeal enhancement varied over time and generally correlated with symptom severity. The appearance was remarkably similar to that of retinal vasculopathy. CONCLUSION: Ce 3D FLAIR may aid in diagnosis and understanding of pathophysiology in Susac's syndrome and may serve as a biomarker for disease activity.


Subject(s)
Image Interpretation, Computer-Assisted , Imaging, Three-Dimensional , Magnetic Resonance Imaging/methods , Meninges/diagnostic imaging , Susac Syndrome/diagnostic imaging , Adult , Contrast Media/administration & dosage , Fluorescein Angiography , Humans , Male , Meninges/drug effects , Predictive Value of Tests , Severity of Illness Index , Steroids/administration & dosage , Susac Syndrome/drug therapy , Time Factors , Treatment Outcome
11.
Cephalalgia ; 36(9): 875-86, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26984967

ABSTRACT

BACKGROUND: Administration of onabotulinumtoxinA (BoNT-A) to peripheral tissues outside the calvaria reduces the number of days chronic migraine patients experience headache. Because the headache phase of a migraine attack, especially those preceded by aura, is thought to involve activation of meningeal nociceptors by endogenous stimuli such as changes in intracranial pressure (i.e. mechanical) or chemical irritants that appear in the meninges as a result of a yet-to-be-discovered sequence of molecular/cellular events triggered by the aura, we sought to determine whether extracranial injections of BoNT-A alter the chemosensitivity of meningeal nociceptors to stimulation of their intracranial receptive fields. MATERIAL AND METHODS: Using electrophysiological techniques, we identified 161 C- and 135 Aδ-meningeal nociceptors in rats and determined their mechanical response threshold and responsiveness to chemical stimulation of their dural receptive fields with TRPV1 and TRPA1 agonists seven days after BoNT-A administration to different extracranial sites. Two paradigms were compared: distribution of 5 U BoNT-A to the lambdoid and sagittal sutures alone, and 1.25 U to the sutures and 3.75 U to the temporalis and trapezius muscles. RESULTS: Seven days after it was administered to tissues outside the calvaria, BoNT-A inhibited responses of C-type meningeal nociceptors to stimulation of their intracranial dural receptive fields with the TRPV1 agonist capsaicin and the TRPA1 agonist mustard oil. BoNT-A inhibition of responses to capsaicin was more effective when the entire dose was injected along the suture lines than when it was injected into muscles and sutures. As in our previous study, BoNT-A had no effect on non-noxious mechanosensitivity of C-fibers or on responsiveness of Aδ-fibers to mechanical and chemical stimulation. DISCUSSION: This study demonstrates that extracranial administration of BoNT-A suppresses meningeal nociceptors' responses to stimulation of their intracranial dural receptive fields with capsaicin and mustard oil. The findings suggest that surface expression of TRPV1 and TRPA1 channels in dural nerve endings of meningeal nociceptors is reduced seven days after extracranial administration of BoNT-A. In the context of chronic migraine, reduced sensitivity to molecules that activate meningeal nociceptors through the TRPV1 and TRPA1 channels can be important for BoNT-A's ability to act as a prophylactic.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Meninges/drug effects , Migraine Disorders/physiopathology , Neuromuscular Agents/pharmacology , Nociceptors/drug effects , Animals , Capsaicin/pharmacology , Cranial Sutures/drug effects , Male , Muscle, Skeletal/drug effects , Mustard Plant , Plant Oils/pharmacology , Rats , Rats, Sprague-Dawley , Sensory System Agents/pharmacology , TRPA1 Cation Channel , TRPC Cation Channels/agonists , TRPV Cation Channels/agonists
12.
Neurobiol Dis ; 79: 111-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25958249

ABSTRACT

Migraine secondary to meningeal input is referred to extracranial regions innervated by somatic afferents that project to homologous regions in the trigeminal nucleus caudalis (TNC). Reported efficacy of extracranial botulinum toxin (BoNT) in treating migraine is surprising since a local extracranial effect of BoNT cannot account for its effect upon meningeal input. We hypothesize that intradermal BoNT acts through central transport in somatic afferents. Anesthetized C57Bl/6 mice (male) received unilateral supraorbital (SO) injections of BoNT-B (1.5 U/40 µl) or saline. 3 days later, mice received ipsilateral (ipsi)-SO capsaicin (20 µl of 0.5mM solution) or meningeal capsaicin (4 µl of 0.35 µM). Pre-treatment with ipsi-SO BoNT-B i) decreased nocicsponsive ipsilateral wiping behavior following ipsi-SO capsaicin; ii) produced cleavage of VAMP in the V1 region of ipsi-TG and in TG neurons showing WGA after SO injection; iii) reduced expression of c-fos in ipsi-TNC following ipsi-SO capsaicin; iv) reduced c-fos activation and NK-1 internalization in ipsi-TNC secondary to ipsi-meningeal capsaicin; and vi) SO WGA did not label dural afferents. We conclude that BoNT-B is taken up by peripheral afferents and transported to central terminals where it inhibits transmitter release resulting in decreased activation of second order neurons. Further, this study supports the hypothesis that SO BoNT exerts a trans-synaptic action on either the second order neuron (which receives convergent input from the meningeal afferent) or the terminal/TG of the converging meningeal afferent.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Membrane Transport Modulators/pharmacology , Migraine Disorders/drug therapy , Neurons, Afferent/drug effects , Trigeminal Caudal Nucleus/drug effects , Afferent Pathways/drug effects , Afferent Pathways/pathology , Afferent Pathways/physiopathology , Animals , Capsaicin , DNA-Binding Proteins , Disease Models, Animal , Face , Male , Meninges/blood supply , Meninges/drug effects , Meninges/pathology , Mice, Inbred C57BL , Migraine Disorders/pathology , Migraine Disorders/physiopathology , Nerve Tissue Proteins/metabolism , Neurons, Afferent/pathology , Neurons, Afferent/physiology , Nociceptive Pain/drug therapy , Nociceptive Pain/pathology , Nociceptive Pain/physiopathology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Neurokinin-1/metabolism , SNARE Proteins/metabolism , Skin/drug effects , Skin/innervation , Trigeminal Caudal Nucleus/pathology , Trigeminal Caudal Nucleus/physiopathology
13.
Acta Neuropathol ; 130(1): 107-18, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869642

ABSTRACT

The Type I interferons (IFN), beta (IFN-ß) and the alpha family (IFN-α), act through a common receptor and have anti-inflammatory effects. IFN-ß is used to treat multiple sclerosis (MS) and is effective against experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Mice with EAE show elevated levels of Type I IFNs in the central nervous system (CNS), suggesting a role for endogenous Type I IFN during inflammation. However, the therapeutic benefit of Type I IFN produced in the CNS remains to be established. The aim of this study was to examine whether experimentally induced CNS-endogenous Type I IFN influences EAE. Using IFN-ß reporter mice, we showed that direct administration of polyinosinic-polycytidylic acid (poly I:C), a potent inducer of IFN-ß, into the cerebrospinal fluid induced increased leukocyte numbers and transient upregulation of IFN-ß in CD45/CD11b-positive cells located in the meninges and choroid plexus, as well as enhanced IFN-ß expression by parenchymal microglial cells. Intrathecal injection of poly I:C to mice showing first symptoms of EAE substantially increased the normal disease-associated expression of IFN-α, IFN-ß, interferon regulatory factor-7 and IL-10 in CNS, and disease worsening was prevented for as long as IFN-α/ß was expressed. In contrast, there was no therapeutic effect on EAE in poly I:C-treated IFN receptor-deficient mice. IFN-dependent microglial and astrocyte response included production of the chemokine CXCL10. These results show that Type I IFN induced within the CNS can play a protective role in EAE and highlight the role of endogenous type I IFN in mediating neuroprotection.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon-alpha/metabolism , Interferon-beta/metabolism , Neuroprotective Agents/pharmacology , Poly I-C/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/pathology , Brain/drug effects , Brain/immunology , Brain/pathology , Chemokine CXCL10/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interferon-alpha/genetics , Interferon-beta/genetics , Leukocytes/drug effects , Leukocytes/pathology , Leukocytes/physiology , Meninges/drug effects , Meninges/immunology , Meninges/pathology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Microglia/physiology , Random Allocation , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology
14.
Ann Neurol ; 73(6): 741-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23447360

ABSTRACT

OBJECTIVE: To examine changes in the response properties of meningeal nociceptors that might lead to migraine pain and examine endogenous processes that could play a role in mediating them using a clinically relevant model of migraine triggering, namely infusion of the nitric oxide (NO) donor nitroglycerin (NTG). METHODS: Single-unit recordings made in the trigeminal ganglion of rats were used to test changes in the activity and mechanosensitivity of meningeal nociceptors in response to administration of the migraine trigger NTG or another NO donor S-nitroso-N-acetyl-DL-penicillamine (SNAP) at doses relevant to the human model of migraine headache. Immunohistochemistry and pharmacological manipulations were used to investigate the possible role of meningeal vascular signaling in mediating the responses of meningeal nociceptors to NO. RESULTS: Infusion of NTG promoted a delayed and robust increase in the mechanosensitivity of meningeal nociceptors, with a time course resembling the development of the delayed migraine headache. A similar sensitization was elicited by dural application of NTG and SNAP. NTG-evoked delayed meningeal nociceptor sensitization was associated with a robust extracellular signal-regulated kinase (ERK) phosphorylation in meningeal arteries. Pharmacological blockade of meningeal ERK phosphorylation inhibited the development of NTG-evoked delayed meningeal nociceptor sensitization. INTERPRETATION: The development of delayed mechanical sensitization evoked by the migraine trigger NTG is potentially of great importance as the first finding of a neurophysiological correlate of migraine headache in meningeal nociceptors. The arterial ERK phosphorylation and its involvement in mediating the NTG-evoked delayed sensitization points to an important, yet unappreciated, role of the meningeal vasculature in the genesis of migraine pain.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Meninges/blood supply , Meninges/enzymology , Migraine Disorders/enzymology , Nociceptors/enzymology , Animals , Male , Meninges/drug effects , Migraine Disorders/chemically induced , Nitric Oxide Donors/toxicity , Nociceptors/drug effects , Rats , Rats, Sprague-Dawley , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/enzymology
15.
Cephalalgia ; 34(11): 853-69, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24694964

ABSTRACT

BACKGROUND: Meningeal and other trigeminal nociceptors are thought to play important roles in the initiation of migraine headache. Currently, the only approved peripherally administered chronic migraine prophylactic drug is onabotulinumtoxinA. The purpose of this study was to determine how botulinum neurotoxin type A (BoNT-A) affects naïve and sensitized meningeal nociceptors. MATERIAL AND METHODS: Using electrophysiological techniques, we identified 43 C- and 36 Aδ-meningeal nociceptors, and measured their spontaneous and evoked firing before and after BoNT-A administration to intracranial dura and extracranial suture-receptive fields. RESULTS: As a rule, BoNT-A inhibited C- but not Aδ-meningeal nociceptors. When applied to nonsensitized C-units, BoNT-A inhibited responses to mechanical stimulation of the dura with suprathreshold forces. When applied to sensitized units, BoNT-A reversed mechanical hypersensitivity. When applied before sensitization, BoNT-A prevented development of mechanical hypersensitivity. When applied extracranially to suture branches of intracranial meningeal nociceptors, BoNT-A inhibited the mechanical responsiveness of the suture branch but not dural axon. In contrast, BoNT-A did not inhibit C-unit responses to mechanical stimulation of the dura with threshold forces, or their spontaneous activity. DISCUSSION: The study provides evidence for the ability of BoNT-A to inhibit mechanical nociception in peripheral trigeminovascular neurons. These findings suggest that BoNT-A interferes with neuronal surface expression of high-threshold mechanosensitive ion channels linked preferentially to mechanical pain by preventing their fusion into the nerve terminal membrane.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Meninges/drug effects , Migraine Disorders/physiopathology , Neurotoxins/pharmacology , Nociceptors/drug effects , Animals , Electrophysiology , Male , Mechanotransduction, Cellular/physiology , Microelectrodes , Pain/physiopathology , Rats , Rats, Sprague-Dawley
16.
Phytomedicine ; 130: 155753, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38795693

ABSTRACT

BACKGROUND: Meningeal lymphatic vessels (mLVs) have great potential to be the therapeutic target for ß Amyloid protein (Aß) clearing in Alzheimer's disease (AD), but the regulatory methods of the mLVs are limited. The lymphatic valve, marked by FOXC2, is the fundamental structure for maintaining stable lymphatic drainage function. Preliminary evidence suggested that borneol (BO) as the classical phytochemicals could enhance the expression of FOXC2 in the mLVs of healthy mice. PURPOSE: This study aims to explore the regulatory ability of BO on lymphatic valves of mLVs in the AD model mice. STUDY DESIGN: We used the intracerebroventricular injection of Aß42 oligomers to construct the AD-like symptoms model induced by toxic protein deposition. We administered BO nano micelles(BO-Ms) orally before and after to simulate the AD prevention and treatment strategy. METHODS: Herein, this study characterized the efficacy and pathways of BO-Ms for regulating mLVs in AD model by Rt-PCR, WB and confocal microscopy, and determined the effects of BO-Ms on Aß clearance, behavior and safety of AD mice. RESULTS: The AD modeling process severely impaired the expression of lymphatic valves. However, after oral administering BO-Ms for prevention and treatment, an increase in the lymphatic valves of the transverse sinus was observed, which derived from the up-regulation of the transcription factor (FOXC2 and Akt) and the down-regulation of the transcription inhibitors (FOXO1 and PRDM1). Furthermore, the effects of BO-Ms on the lymphatic valves could enhance the lymphatic drainage of the mLVs in AD-like mice, promoting the clearance of toxicity aggregates, protecting neurons, and alleviating AD-like symptoms. Simultaneously, continuous oral BO-Ms for 30 days didn't show any significant organ toxicity. The most important thing was that the preventive effect of BO administration was superior to therapeutic administration in all data. CONCLUSION: In summary, our research indicated that BO is a promoter of lymphatic valve formation in the mLVs, and could prevent or repair damage caused by toxic Aß42. BO was the only bioactive natural product with the ability to regulate mLVs valves. Thus, BO has the potential to become phytochemicals for alleviating AD symptoms by enhancing the drainage function of mLVs.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Camphanes , Disease Models, Animal , Forkhead Transcription Factors , Animals , Amyloid beta-Peptides/metabolism , Alzheimer Disease/prevention & control , Alzheimer Disease/drug therapy , Mice , Camphanes/pharmacology , Forkhead Transcription Factors/metabolism , Male , Lymphatic Vessels/drug effects , Meninges/drug effects , Mice, Inbred C57BL
17.
Theranostics ; 14(11): 4331-4351, 2024.
Article in English | MEDLINE | ID: mdl-39113801

ABSTRACT

Background: The impediment to ß-amyloid (Aß) clearance caused by the invalid intracranial lymphatic drainage in Alzheimer's disease is pivotal to its pathogenesis, and finding reliable clinical available solutions to address this challenge remains elusive. Methods: The potential role and underlying mechanisms of intranasal oxytocin administration, an approved clinical intervention, in improving intracranial lymphatic drainage in middle-old-aged APP/PS1 mice were investigated by live mouse imaging, ASL/CEST-MRI scanning, in vivo two-photon imaging, immunofluorescence staining, ELISA, RT-qPCR, Western blotting, RNA-seq analysis, and cognitive behavioral tests. Results: Benefiting from multifaceted modulation of cerebral hemodynamics, aquaporin-4 polarization, meningeal lymphangiogenesis and transcriptional profiles, oxytocin administration normalized the structure and function of both the glymphatic and meningeal lymphatic systems severely impaired in middle-old-aged APP/PS1 mice. Consequently, this intervention facilitated the efficient drainage of Aß from the brain parenchyma to the cerebrospinal fluid and then to the deep cervical lymph nodes for efficient clearance, as well as improvements in cognitive deficits. Conclusion: This work broadens the underlying neuroprotective mechanisms and clinical applications of oxytocin medication, showcasing its promising therapeutic prospects in central nervous system diseases with intracranial lymphatic dysfunction.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Glymphatic System , Mice, Transgenic , Oxytocin , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Mice , Oxytocin/pharmacology , Oxytocin/administration & dosage , Oxytocin/metabolism , Glymphatic System/metabolism , Glymphatic System/drug effects , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/drug effects , Brain/diagnostic imaging , Administration, Intranasal , Lymphangiogenesis/drug effects , Male , Aquaporin 4/metabolism , Aquaporin 4/genetics , Humans , Magnetic Resonance Imaging , Meninges/metabolism , Meninges/drug effects , Meninges/diagnostic imaging
18.
Dev Biol ; 369(1): 101-14, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22771246

ABSTRACT

Embryonic development is controlled by a small set of signal transduction pathways, with vastly different phenotypic outcomes depending on the time and place of their recruitment. How the same molecular machinery can elicit such specific and distinct responses, remains one of the outstanding questions in developmental biology. Part of the answer may lie in the high inherent genetic complexity of these signaling cascades, as observed for the Wnt-pathway. The mammalian genome encodes multiple Wnt proteins and receptors, each of which show dynamic and tightly controlled expression patterns in the embryo. Yet how these components interact in the context of the whole organism remains unknown. Here we report the generation of a novel, inducible transgenic mouse model that allows spatiotemporal control over the expression of Wnt5a, a protein implicated in many developmental processes and multiple Wnt-signaling responses. We show that ectopic Wnt5a expression from E10.5 onwards results in a variety of developmental defects, including loss of hair follicles and reduced bone formation in the skull. Moreover, we find that Wnt5a can have dual signaling activities during mouse embryonic development. Specifically, Wnt5a is capable of both inducing and repressing ß-catenin/TCF signaling in vivo, depending on the time and site of expression and the receptors expressed by receiving cells. These experiments show for the first time that a single mammalian Wnt protein can have multiple signaling activities in vivo, thereby furthering our understanding of how signaling specificity is achieved in a complex developmental context.


Subject(s)
Embryonic Development/genetics , Repressor Proteins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Animals , Bone and Bones/abnormalities , Bone and Bones/drug effects , Bone and Bones/embryology , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Hair Follicle/cytology , Hair Follicle/drug effects , Hair Follicle/embryology , Intercellular Signaling Peptides and Proteins/metabolism , Meninges/drug effects , Meninges/embryology , Mice , Mice, Transgenic , Models, Animal , Osteogenesis/drug effects , Phenotype , Pregnancy , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Skin/drug effects , Skin/embryology , Skin/metabolism , Tetracycline/pharmacology , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects , Wnt-5a Protein , beta Catenin/metabolism
19.
BMC Genomics ; 14: 147, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23497014

ABSTRACT

BACKGROUND: The meninges (arachnoid and pial membranes) and associated vasculature (MAV) and choroid plexus are important in maintaining cerebrospinal fluid (CSF) generation and flow. MAV vasculature was previously observed to be adversely affected by environmentally-induced hyperthermia (EIH) and more so by a neurotoxic amphetamine (AMPH) exposure. Herein, microarray and RT-PCR analysis was used to compare the gene expression profiles between choroid plexus and MAV under control conditions and at 3 hours and 1 day after EIH or AMPH exposure. Since AMPH and EIH are so disruptive to vasculature, genes related to vasculature integrity and function were of interest. RESULTS: Our data shows that, under control conditions, many of the genes with relatively high expression in both the MAV and choroid plexus are also abundant in many epithelial tissues. These genes function in transport of water, ions, and solutes, and likely play a role in CSF regulation. Most genes that help form the blood-brain barrier (BBB) and tight junctions were also highly expressed in MAV but not in choroid plexus. In MAV, exposure to EIH and more so to AMPH decreased the expression of BBB-related genes such as Sox18, Ocln, and Cldn5, but they were much less affected in the choroid plexus. There was a correlation between the genes related to reactive oxidative stress and damage that were significantly altered in the MAV and choroid plexus after either EIH or AMPH. However, AMPH (at 3 hr) significantly affected about 5 times as many genes as EIH in the MAV, while in the choroid plexus EIH affected more genes than AMPH. Several unique genes that are not specifically related to vascular damage increased to a much greater extent after AMPH compared to EIH in the MAV (Lbp, Reg3a, Reg3b, Slc15a1, Sct and Fst) and choroid plexus (Bmp4, Dio2 and Lbp). CONCLUSIONS: Our study indicates that the disruption of choroid plexus function and damage produced by AMPH and EIH is significant, but the changes may not be as pronounced as they are in the MAV, particularly for AMPH. Expression profiles in the MAV and choroid plexus differed to some extent and differences were not restricted to vascular related genes.


Subject(s)
Brain/metabolism , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Meninges/metabolism , Amphetamine/toxicity , Arachnoid/blood supply , Arachnoid/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/blood supply , Choroid Plexus/blood supply , Choroid Plexus/drug effects , Environment , Fever , Humans , Meninges/blood supply , Meninges/drug effects , Pancreatitis-Associated Proteins , Transcriptome
20.
Antimicrob Agents Chemother ; 57(12): 5808-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24002097

ABSTRACT

In this study, the efficacy of ceftaroline fosamil was compared with that of cefepime in an experimental rabbit meningitis model against two Gram-negative strains (Escherichia coli QK-9 and Klebsiella pneumoniae 1173687). The penetration of ceftaroline into inflamed and uninflamed meninges was also investigated. Both regimens were bactericidal, but ceftaroline fosamil was significantly superior to cefepime against K. pneumoniae and E. coli in this experimental rabbit meningitis model (P < 0.0007 against K. pneumoniae and P < 0.0016 against E. coli). The penetration of ceftaroline was approximately 15% into inflamed meninges and approximately 3% into uninflamed meninges.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Klebsiella Infections/drug therapy , Meningitis, Escherichia coli/drug therapy , Animals , Anti-Bacterial Agents/cerebrospinal fluid , Anti-Bacterial Agents/pharmacokinetics , Cefepime , Cephalosporins/cerebrospinal fluid , Cephalosporins/pharmacokinetics , Disease Models, Animal , Escherichia coli/drug effects , Escherichia coli/physiology , Klebsiella Infections/cerebrospinal fluid , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/physiology , Meninges/drug effects , Meninges/metabolism , Meninges/microbiology , Meningitis, Escherichia coli/cerebrospinal fluid , Meningitis, Escherichia coli/microbiology , Permeability , Rabbits , Treatment Outcome , Ceftaroline
SELECTION OF CITATIONS
SEARCH DETAIL