Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 185(26): 5040-5058.e19, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563667

ABSTRACT

Spatial molecular profiling of complex tissues is essential to investigate cellular function in physiological and pathological states. However, methods for molecular analysis of large biological specimens imaged in 3D are lacking. Here, we present DISCO-MS, a technology that combines whole-organ/whole-organism clearing and imaging, deep-learning-based image analysis, robotic tissue extraction, and ultra-high-sensitivity mass spectrometry. DISCO-MS yielded proteome data indistinguishable from uncleared samples in both rodent and human tissues. We used DISCO-MS to investigate microglia activation along axonal tracts after brain injury and characterized early- and late-stage individual amyloid-beta plaques in a mouse model of Alzheimer's disease. DISCO-bot robotic sample extraction enabled us to study the regional heterogeneity of immune cells in intact mouse bodies and aortic plaques in a complete human heart. DISCO-MS enables unbiased proteome analysis of preclinical and clinical tissues after unbiased imaging of entire specimens in 3D, identifying diagnostic and therapeutic opportunities for complex diseases. VIDEO ABSTRACT.


Subject(s)
Alzheimer Disease , Proteome , Mice , Humans , Animals , Proteome/analysis , Proteomics/methods , Alzheimer Disease/pathology , Amyloid beta-Peptides , Mass Spectrometry , Plaque, Amyloid
2.
Cereb Cortex ; 33(4): 1074-1089, 2023 02 07.
Article in English | MEDLINE | ID: mdl-35353195

ABSTRACT

At present, many studies support the notion that after stroke, remote regions connected to the infarcted area are also affected and may contribute to functional outcome. In the present study, we have analyzed possible microanatomical alterations in pyramidal neurons from the contralesional hemisphere after induced stroke. We performed intracellular injections of Lucifer yellow in pyramidal neurons from layer III in the somatosensory cortex of the contralesional hemisphere in an ischemic stroke mouse model. A detailed 3-dimensional analysis of the neuronal complexity and morphological alterations of dendritic spines was then performed. Our results demonstrate that pyramidal neurons from layer III in the somatosensory cortex of the contralesional hemisphere show selective changes in their dendritic arbors, namely, less dendritic complexity of the apical dendritic arbor-but no changes in the basal dendritic arbor. In addition, we found differences in spine morphology in both apical and basal dendrites comparing the contralesional hemisphere with the lesional hemisphere. Our results show that pyramidal neurons of remote areas connected to the infarct zone exhibit a series of selective changes in neuronal complexity and morphological distribution of dendritic spines, supporting the hypothesis that remote regions connected to the peri-infarcted area are also affected after stroke.


Subject(s)
Ischemic Stroke , Stroke , Mice , Animals , Somatosensory Cortex , Pyramidal Cells/physiology , Neurons , Dendrites/physiology
3.
Medicina (Kaunas) ; 59(12)2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38138271

ABSTRACT

Background and Objectives: Dissecting the complex pathological cascade of an ischemic stroke in preclinical models is highly warranted to understand the course of this disease in humans. Neurogenesis and angiogenesis are integral for post-stroke recovery, yet it is not clear how these processes are altered months after an ischemic stroke. In this study, we investigated the changes that take place subacutely after focal cerebral ischemia in experimental adult male mice. Materials and Methods: Male 12-week-old C57BL/6 mice underwent a 60 min long fMCAo or sham surgery. Two months after the procedure, we examined the immunohistochemistry to assess the changes in neuroblast (DCX) and differentiated neuron (NeuN) numbers, as well as the density of the pro-angiogenic factor VEGF. Results: We found decreased neuroblast numbers in both brain hemispheres of the fMCAo mice: by more than 85% in the dentate gyrus and by more than 70% in the subventricular zone. No neuroblasts were found in the contralateral hemisphere of the fMCAO mice or the sham controls, but a small population was detected in the ipsilateral ischemic core of the fMCAo mice. Intriguingly, the number of differentiated neurons in the ipsilateral ischemic core was lower by 20% compared to the contralateral hemisphere. VEGF expression was diminished in both brain hemispheres of the fMCAo mice. Conclusions: Our current report shows that focal cerebral ischemia induces changes in neuroblast numbers and the pro-angiogenic factor VEGF in both cerebral hemispheres 2 months after an fMCAo in mice. Our data show that focal cerebral ischemia induces a long-term regenerative response in both brain hemispheres.


Subject(s)
Brain Ischemia , Ischemic Stroke , Humans , Mice , Male , Animals , Angiogenesis Inducing Agents , Vascular Endothelial Growth Factor A/metabolism , Mice, Inbred C57BL , Brain Ischemia/complications , Neurons/metabolism , Cerebral Infarction/pathology , Ischemia/pathology
4.
Nanomedicine ; 40: 102511, 2022 02.
Article in English | MEDLINE | ID: mdl-34915181

ABSTRACT

The potential of poly(lactic-co-glycolic acid) (PLGA) to design nanoparticles (NPs) and target the central nervous system remains to be exploited. In the current study we designed fluorescent 70-nm PLGA NPs, loaded with bulky fluorophores, thereby making them significantly brighter than quantum dots in single-particle fluorescence measurements. The high brightness of NPs enabled their visualization by intravital real-time 2-photon microscopy. Subsequently, we found that PLGA NPs coated with pluronic F-68 circulated in the blood substantially longer than uncoated NPs and were taken up by cerebro-vascular endothelial cells. Additionally, confocal microscopy revealed that coated PLGA NPs were present in late endothelial endosomes of cerebral vessels within 1 h after systemic injection and were more readily taken up by endothelial cells in peripheral organs. The combination of ultra-bright NPs and in vivo imaging may thus represent a promising approach to reduce the gap between development and clinical application of nanoparticle-based drug carriers.


Subject(s)
Nanoparticles , Poloxamer , Drug Carriers , Endothelial Cells , Glycols , Microscopy , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer
5.
Nat Methods ; 13(10): 859-67, 2016 10.
Article in English | MEDLINE | ID: mdl-27548807

ABSTRACT

Recent tissue-clearing approaches have become important alternatives to standard histology approaches. However, light scattering in thick tissues and the size restrictions on samples that can be imaged with standard light-sheet microscopy pose limitations for analyzing large samples such as an entire rodent body. We developed 'ultimate DISCO' (uDISCO) clearing to overcome these limitations in volumetric imaging. uDISCO preserves fluorescent proteins over months and renders intact organs and rodent bodies transparent while reducing their size up to 65%. We used uDISCO to image neuronal connections and vasculature from head to toe over 7 cm and to perform unbiased screening of transplanted stem cells within the entire body of adult mice. uDISCO is compatible with diverse labeling methods and archival human tissue, and it can readily be used in various biomedical applications to study organization of large organ systems throughout entire organisms.


Subject(s)
Imaging, Three-Dimensional/methods , Neuroimaging/methods , Single-Cell Analysis/methods , Whole Body Imaging/methods , Animals , Central Nervous System/blood supply , Central Nervous System/cytology , Contrast Media , Female , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Half-Life , Humans , Immunohistochemistry/methods , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence/methods , Organ Specificity , Phenyl Ethers/chemistry , Rats , Solvents/chemistry , Staining and Labeling
6.
Ann Neurol ; 78(6): 887-900, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26312599

ABSTRACT

OBJECTIVE: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most common inherited small-vessel disease, is associated with vascular aggregation of mutant Notch3 protein, dysfunction of cerebral vessels, and dementia. Pericytes, perivascular cells involved in microvascular function, express Notch3. Therefore, we hypothesize that these cells may play a role in the pathogenesis of CADASIL. METHODS: Two-, 7-, and 12-month-old CADASIL mutant mice (TgNotch3(R169C) ) and wild-type controls were examined regarding Notch3 aggregation in pericytes, the coverage of cerebral vessels by pericytes, pericyte numbers, capillary density, blood-brain barrier (BBB) integrity, astrocytic end-feet, and the expression of astrocytic gap junction and endothelial adherens junction protein using immunostaining and Western blot analysis. In addition, we examined cerebrovascular CO2 reactivity using laser Doppler fluxmetry and in vivo microscopy. RESULTS: With increasing age, mutated Notch3 aggregated around pericytes and smooth muscle cells. Notch3 aggregation caused significant reduction of pericyte number and coverage of capillaries by pericyte processes (p < 0.01). These changes were associated with detachment of astrocytic end-feet from cerebral microvessels, leakage of plasma proteins, reduction in expression of endothelial adherens junction protein, and reduced microvascular reactivity to CO2 . Smooth muscle cells were not affected by Notch3 accumulation. INTERPRETATION: Our results show that pericytes are the first cells affected by Notch3 aggregation in CADASIL mice. Pericyte pathology causes opening of the BBB and microvascular dysfunction. Therefore, protecting pericytes may represent a novel therapeutic strategy for vascular dementia.


Subject(s)
Blood-Brain Barrier/pathology , CADASIL/etiology , Capillaries/pathology , Cerebral Cortex/blood supply , Pericytes/pathology , Receptors, Notch/metabolism , Age Factors , Animals , CADASIL/metabolism , CADASIL/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Mutation , Pericytes/metabolism , Random Allocation , Receptor, Notch3 , Receptors, Notch/genetics , Single-Blind Method
7.
Brain Pathol ; 34(3): e13222, 2024 May.
Article in English | MEDLINE | ID: mdl-38012061

ABSTRACT

Significant progress has been made with regard to understanding how the adult brain responds after a stroke. However, a large number of patients continue to suffer lifelong disabilities without adequate treatment. In the present study, we have analyzed possible microanatomical alterations in the contralesional hippocampus from the ischemic stroke mouse model tMCAo 12-14 weeks after transient middle cerebral artery occlusion. After individually injecting Lucifer yellow into pyramidal neurons from the CA1 field of the hippocampus, we performed a detailed three-dimensional analysis of the neuronal complexity, dendritic spine density, and morphology. We found that, in both apical (stratum radiatum) and basal (stratum oriens) arbors, CA1 pyramidal neurons in the contralesional hippocampus of tMCAo mice have a significantly higher neuronal complexity, as well as reduced spine density and alterations in spine volume and spine length. Our results show that when the ipsilateral hippocampus is dramatically damaged, the contralesional hippocampus exhibits several statistically significant selective alterations. However, these alterations are not as significant as expected, which may help to explain the recovery of hippocampal function after stroke. Further anatomical and physiological studies are necessary to better understand the modifications in the "intact" contralesional lesioned brain regions, which are probably fundamental to recover functions after stroke.


Subject(s)
Hippocampus , Pyramidal Cells , Humans , Mice , Animals , CA1 Region, Hippocampal , Neurons , Infarction, Middle Cerebral Artery , Dendritic Spines , Dendrites
8.
Nat Biotechnol ; 42(4): 617-627, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37430076

ABSTRACT

Whole-body imaging techniques play a vital role in exploring the interplay of physiological systems in maintaining health and driving disease. We introduce wildDISCO, a new approach for whole-body immunolabeling, optical clearing and imaging in mice, circumventing the need for transgenic reporter animals or nanobody labeling and so overcoming existing technical limitations. We identified heptakis(2,6-di-O-methyl)-ß-cyclodextrin as a potent enhancer of cholesterol extraction and membrane permeabilization, enabling deep, homogeneous penetration of standard antibodies without aggregation. WildDISCO facilitates imaging of peripheral nervous systems, lymphatic vessels and immune cells in whole mice at cellular resolution by labeling diverse endogenous proteins. Additionally, we examined rare proliferating cells and the effects of biological perturbations, as demonstrated in germ-free mice. We applied wildDISCO to map tertiary lymphoid structures in the context of breast cancer, considering both primary tumor and metastases throughout the mouse body. An atlas of high-resolution images showcasing mouse nervous, lymphatic and vascular systems is accessible at http://discotechnologies.org/wildDISCO/atlas/index.php .


Subject(s)
Imaging, Three-Dimensional , Immunoglobulin G , Mice , Animals
9.
J Neurosci Methods ; 372: 109493, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35151669

ABSTRACT

INTRODUCTION: Histology on fixed brain tissue is a key technique to investigate the pathophysiology of neurological disorders. Best results are obtained by perfusion fixation, however, multiple protocols are available and so far the optimal perfusion pressure (PP) for the preservation of brain tissue while also maintaining vascular integrity is not defined. Therefore, the aim of our study was to investigate the effect of different PPs on the cerebral vasculature and to define the PP optimal for the preservation of both vascular integrity and tissue fixation. MATERIAL AND METHODS: Male C57Bl6 mice, 8 weeks old, were perfused with PPs of 50/125/300 mmHg (series I) or 50/100/150/300 mmHg (series II). In series I, vascular integrity, e.g. BBB permeability, vessel diameter, and occurrence of vasospasms were investigated by spectrophotometry, light-sheet and 2-photon microscopy, respectively. In series II, we investigated vascular and neuronal artifacts and the occurrence of hemorrhage or microthrombi by light microscopy. RESULTS: While a PP below the physiological systolic blood pressure results in the collapse of parenchymal vessels and formation of microvasospasms and microclots, a PP above the physiological systolic blood pressure dilates cerebral vessels, induces microvasospasms and disrupts the BBB. In terms of tissue integrity, our results confirm that higher PPs lead to fewer artifacts such as dark neurons or perivascular courts. CONCLUSION: Our study demonstrates that the PP critically affects both vascular and tissue integrity in brain tissue preserved by perfusion fixation. A PP between 125 and 150 mmHg is optimal for the preservation of the cerebral vasculature and neuronal structures.


Subject(s)
Brain , Neurons , Animals , Blood-Brain Barrier , Brain/pathology , Male , Mice , Mice, Inbred C57BL , Perfusion/methods , Tissue Fixation/methods
10.
J Neurosci ; 30(20): 6930-43, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484635

ABSTRACT

Neuronal migration and axon growth, key events during neuronal development, require distinct changes in the cytoskeleton. Although many molecular regulators of polarity have been identified and characterized, relatively little is known about their physiological role in this process. To study the physiological function of Rac1 in neuronal development, we have generated a conditional knock-out mouse, in which Rac1 is ablated in the whole brain. Rac1-deficient cerebellar granule neurons, which do not express other Rac isoforms, showed impaired neuronal migration and axon formation both in vivo and in vitro. In addition, Rac1 ablation disrupts lamellipodia formation in growth cones. The analysis of Rac1 effectors revealed the absence of the Wiskott-Aldrich syndrome protein (WASP) family verprolin-homologous protein (WAVE) complex from the plasma membrane of knock-out growth cones. Loss of WAVE function inhibited axon growth, whereas overexpression of a membrane-tethered WAVE mutant partially rescued axon growth in Rac1-knock-out neurons. In addition, pharmacological inhibition of the WAVE complex effector Arp2/3 also reduced axon growth. We propose that Rac1 recruits the WAVE complex to the plasma membrane to enable actin remodeling necessary for axon growth.


Subject(s)
Cell Movement/physiology , Neurons/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac1 GTP-Binding Protein/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Bromodeoxyuridine/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cofilin 1/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Growth Cones/drug effects , Growth Cones/metabolism , Ki-67 Antigen/metabolism , Luminescent Proteins/genetics , Mice , Mice, Knockout , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Organ Culture Techniques/methods , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Transfection/methods , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/metabolism
11.
Acta Neuropathol Commun ; 9(1): 138, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404478

ABSTRACT

Traumatic brain injury (TBI) causes acute and subacute tissue damage, but is also associated with chronic inflammation and progressive loss of brain tissue months and years after the initial event. The trigger and the subsequent molecular mechanisms causing chronic brain injury after TBI are not well understood. The aim of the current study was therefore to investigate the hypothesis that necroptosis, a form a programmed cell death mediated by the interaction of Receptor Interacting Protein Kinases (RIPK) 1 and 3, is involved in this process. Neuron-specific RIPK1- or RIPK3-deficient mice and their wild-type littermates were subjected to experimental TBI by controlled cortical impact. Posttraumatic brain damage and functional outcome were assessed longitudinally by repetitive magnetic resonance imaging (MRI) and behavioral tests (beam walk, Barnes maze, and tail suspension), respectively, for up to three months after injury. Thereafter, brains were investigated by immunohistochemistry for the necroptotic marker phosphorylated mixed lineage kinase like protein(pMLKL) and activation of astrocytes and microglia. WT mice showed progressive chronic brain damage in cortex and hippocampus and increased levels of pMLKL after TBI. Chronic brain damage occurred almost exclusively in areas with iron deposits and was significantly reduced in RIPK1- or RIPK3-deficient mice by up to 80%. Neuroprotection was accompanied by a reduction of astrocyte and microglia activation and improved memory function. The data of the current study suggest that progressive chronic brain damage and cognitive decline after TBI depend on the expression of RIPK1/3 in neurons. Hence, inhibition of necroptosis signaling may represent a novel therapeutic target for the prevention of chronic post-traumatic brain damage.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/genetics , Brain/metabolism , Microglia/metabolism , Necroptosis/genetics , Neurons/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Brain/diagnostic imaging , Brain/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Brain Injury, Chronic/genetics , Brain Injury, Chronic/metabolism , Brain Injury, Chronic/pathology , Brain Injury, Chronic/physiopathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hindlimb Suspension , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/pathology , Magnetic Resonance Imaging , Maze Learning , Memory , Mice , Mice, Knockout , Neurons/pathology , Protein Kinases/metabolism
12.
ACS Nano ; 14(8): 9755-9770, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32680421

ABSTRACT

Visualizing single organic nanoparticles (NPs) in vivo remains a challenge, which could greatly improve our understanding of the bottlenecks in the field of nanomedicine. To achieve high single-particle fluorescence brightness, we loaded polymer poly(methyl methacrylate)-sulfonate (PMMA-SO3H) NPs with octadecyl rhodamine B together with a bulky hydrophobic counterion (perfluorinated tetraphenylborate) as a fluorophore insulator to prevent aggregation-caused quenching. To create NPs with stealth properties, we used the amphiphilic block copolymers pluronic F-127 and F-68. Fluorescence correlation spectroscopy and Förster resonance energy transfer (FRET) revealed that pluronics remained at the NP surface after dialysis (at one amphiphile per 5.5 nm2) and prevented NPs from nonspecific interactions with serum proteins and surfactants. In primary cultured neurons, pluronics stabilized the NPs, preventing their prompt aggregation and binding to neurons. By increasing dye loading to 20 wt % and optimizing particle size, we obtained 74 nm NPs showing 150-fold higher single-particle brightness with two-photon excitation than commercial Nile Red-loaded FluoSpheres of 39 nm hydrodynamic diameter. The obtained ultrabright pluronic-coated NPs enabled direct single-particle tracking in vessels of mice brains by two-photon intravital microscopy for at least 1 h, whereas noncoated NPs were rapidly eliminated from the circulation. Following brain injury or neuroinflammation, which can open the blood-brain barrier, extravasation of NPs was successfully monitored. Moreover, we demonstrated tracking of individual NPs from meningeal vessels until their uptake by meningeal macrophages. Thus, single NPs can be tracked in animals in real time in vivo in different brain compartments and their dynamics visualized with subcellular resolution.


Subject(s)
Nanoparticles , Poloxamer , Animals , Brain , Fluorescent Dyes , Mice , Particle Size , Polymers
13.
J Neurotrauma ; 37(11): 1331-1341, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31847690

ABSTRACT

Increasing clinical and experimental evidence suggests that traumatic brain injury (TBI) is associated with progressive histopathological damage. The aim of the current study was to characterize the time course of motor function, memory performance, and depression-like behavior up to 1 year after experimental TBI, and to correlate these changes to histopathological outcome. Male C57BL/6N mice underwent controlled cortical impact (CCI) or sham operation, and histopathological outcome was evaluated 15 min, 24 h, 1 week, or 1, 3, 6, or 12 months thereafter (n = 12 animals per time point). Motor function, depression-like behavior, and memory function were evaluated concomitantly, and magnetic resonance imaging (MRI) was repeatedly performed. Naïve mice (n = 12) served as an unhandled control group. Injury volume almost doubled within 1 year after CCI (p = 0.008) and the ipsilateral hemisphere became increasingly atrophic (p < 0.0001). Progressive tissue loss was observed in the corpus callosum (p = 0.007) and the hippocampus (p = 0.004) together with hydrocephalus formation (p < 0.0001). Motor function recovered partially after TBI, but 6 months after injury progressive depression-like behavior (p < 0.0001) and loss of memory function (p < 0.0001) were observed. The present study demonstrates that delayed histopathological damage that occurs over months after brain injury is followed by progressive depression and memory loss, changes also observed after TBI in humans. Hence, experimental TBI models in mice replicate long-term sequelae of brain injury such as post-traumatic dementia and depression.


Subject(s)
Brain Injuries, Traumatic/pathology , Cognitive Dysfunction/pathology , Depression/pathology , Disease Progression , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Depression/diagnostic imaging , Depression/etiology , Magnetic Resonance Imaging/trends , Memory Disorders/diagnostic imaging , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Time Factors
14.
J Neurosci ; 27(34): 9169-80, 2007 Aug 22.
Article in English | MEDLINE | ID: mdl-17715353

ABSTRACT

Axons in the CNS do not regrow after injury, whereas lesioned axons in the peripheral nervous system (PNS) regenerate. Lesioned CNS axons form characteristic swellings at their tips known as retraction bulbs, which are the nongrowing counterparts of growth cones. Although much progress has been made in identifying intracellular and molecular mechanisms that regulate growth cone locomotion and axonal elongation, a comprehensive understanding of how retraction bulbs form and why they are unable to grow is still elusive. Here we report the analysis of the morphological and intracellular responses of injured axons in the CNS compared with those in the PNS. We show that retraction bulbs of injured CNS axons increase in size over time, whereas growth cones of injured PNS axons remain constant. Retraction bulbs contain a disorganized microtubule network, whereas growth cones possess the typical bundling of microtubules. Using in vivo imaging, we find that pharmacological disruption of microtubules in growth cones transforms them into retraction bulb-like structures whose growth is inhibited. Correspondingly, microtubule destabilization of sensory neurons in cell culture induces retraction bulb formation. Conversely, microtubule stabilization prevents the formation of retraction bulbs and decreases axonal degeneration in vivo. Finally, microtubule stabilization enhances the growth capacity of CNS neurons cultured on myelin. Thus, the stability and organization of microtubules define the fate of lesioned axonal stumps to become either advancing growth cones or nongrowing retraction bulbs. Our data pinpoint microtubules as a key regulatory target for axonal regeneration.


Subject(s)
Axons/physiology , Microtubules/physiology , Nerve Degeneration/pathology , Regeneration/physiology , Animals , Axons/drug effects , Axons/ultrastructure , Cells, Cultured , Central Nervous System/injuries , Central Nervous System/pathology , Cerebellum/cytology , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Growth Cones/drug effects , Growth Cones/physiology , Growth Cones/ultrastructure , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Nerve Degeneration/etiology , Neurons/cytology , Neurons/metabolism , Neurons/ultrastructure , Nocodazole/pharmacology , Paclitaxel/pharmacology , Sciatic Neuropathy/complications , Tubulin Modulators/pharmacology
15.
J Cereb Blood Flow Metab ; 37(6): 2084-2097, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27449604

ABSTRACT

Experimental stroke models producing clinically relevant functional deficits are often associated with high mortality. Because the mechanisms that underlie post-stroke mortality are largely unknown, results obtained using these models are often difficult to interpret, thereby limiting their translational potential. Given that specific forms of post-stroke care reduce mortality in patients, we hypothesized that inadequate food and water intake may underlie mortality following experimental stroke. C57BL/6 mice were subjected to 1 h of intraluminal filament middle cerebral artery occlusion. Nutritional support beginning on the second day after filament middle cerebral artery occlusion reduced the 14-day mortality rate from 59% to 15%. The surviving mice in the post-stroke support group had the same infarct size as non-surviving control mice, suggesting that post-stroke care was not neuroprotective and that inadequate food and/or water intake are the main reasons for filament middle cerebral artery occlusion-induced mortality. This notion was supported by the presence of significant hypoglycemia, ketonemia, and dehydration in control mice. Taken together, these data suggest that post-filament middle cerebral artery occlusion mortality in mice is not primarily caused by ischemic brain damage, but secondarily by inadequate food and/or water intake. Thus, providing nutritional support following filament middle cerebral artery occlusion greatly minimizes mortality bias and allows the study of long-term morphological and functional sequelae of stroke in mice.


Subject(s)
Drinking/physiology , Eating/physiology , Nutritional Support , Stroke/physiopathology , Stroke/therapy , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL , Survival Analysis
16.
J Neurotrauma ; 23(5): 696-707, 2006 May.
Article in English | MEDLINE | ID: mdl-16689671

ABSTRACT

The potent non-peptide B2 receptor (R) antagonist, Anatibant mesylate (Ms) (LF 16-0687 Ms), reduces brain edema and improves neurological function recovery in various focal and diffuse models of traumatic brain injury in rodents. In the present study, alteration of kinin B1 and B2R after closed head trauma (CHT) and in vivo binding properties of Anatibant Ms (3 mg/kg, s.c.) injected 30 min after CHT were studied in mice by autoradiography using the radioligands [125I]HPP-Hoe 140 (B2R), and [125I]HPP-des-Arg10-Hoe 140 (B1R). Whereas B1R is barely detected in most brain regions, B2R is extensively distributed, displaying the highest densities in the hindbrain. CHT was associated with a slight increase of B1R and a decrease of B2R (10-50%) in several brain regions. Anatibant Ms (Ki = 22 pM) displaced the B2R radioligand from its binding sites in several areas of the forebrain, basal ganglia and hindbrain. Displacement was achieved in 1 h and persisted at 4 h post-injection. The inhibition did not exceed 50% of the total specific binding in non-injured mice. After CHT, the displacement by Anatibant Ms was higher and almost complete in the cortex, caudate putamen, thalamus, hippocampus, medial geniculate nucleus, ventral tegmental area, and raphe. Evans blue extravasation in brain tissue at 4 h after CHT was abolished by Anatibant Ms. It appeared that Anatibant Ms penetrated into the brain in sufficient amounts, particularly after disruption of the blood-brain barrier, to account for its B2R-mediated neuro- and vascular protective effects. The diminished binding of B2R after CHT may reflect the occupancy or internalization of B2R following the endogenous production of bradykinin (BK).


Subject(s)
Blood-Brain Barrier/drug effects , Brain/metabolism , Head Injuries, Closed/physiopathology , Quinolines/pharmacology , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism , Animals , Autoradiography , Binding, Competitive , Blood-Brain Barrier/physiology , Brain/drug effects , Male , Mice , Phenols/pharmacokinetics , Propanols/pharmacokinetics
17.
Front Cell Neurosci ; 9: 54, 2015.
Article in English | MEDLINE | ID: mdl-25755635

ABSTRACT

Neuroinflammation, the inflammatory response in the central nervous system (CNS), is a major determinant of neuronal function and survival during aging and disease progression. Microglia, as the resident tissue-macrophages of the brain, provide constant support to surrounding neurons in healthy brain. Upon any stress signal (such as trauma, ischemia, inflammation) they are one of the first cells to react. Local and/or peripheral signals determine microglia stress response, which can vary within a continuum of states from beneficial to detrimental for neuronal survival, and can be shaped by aging and previous insults. In this review, we discuss the roles of microglia upon an ischemic or traumatic injury, and give our perspective how aging may contribute to microglia behavior in the injured brain. We speculate that a deeper understanding of specific microglia identities will pave the way to develop more potent therapeutics to treat the diseases of aging brain.

18.
J Cereb Blood Flow Metab ; 35(9): 1445-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26058694

ABSTRACT

Aging leads to a gradual decline in the fidelity of cerebral blood flow (CBF) responses to neuronal activation, resulting in an increased risk for stroke and dementia. However, it is currently unknown when age-related cerebrovascular dysfunction starts or which vascular components and functions are first affected. The aim of this study was to examine the function of microcirculation throughout aging in mice. Microcirculation was challenged by inhalation of 5% and 10% CO2 or by forepaw stimulation in 6-week, 8-month, and 12-month-old FVB/N mice. The resulting dilation of pial vessels and increase in CBF was measured by intravital fluorescence microscopy and laser Doppler fluxmetry, respectively. Neurovascular coupling and astrocytic endfoot Ca(2+) were measured in acute brain slices from 18-month-old mice. We did not reveal any changes in CBF after CO2 reactivity up to an age of 12 months. However, direct visualization of pial vessels by in vivo microscopy showed a significant, age-dependent loss of CO2 reactivity starting at 8 months of age. At the same age neurovascular coupling was also significantly affected. These results suggest that aging does not affect cerebral vessel function simultaneously, but starts in pial microvessels months before global changes in CBF are detectable.


Subject(s)
Aging/metabolism , Astrocytes/metabolism , Cerebral Arteries/physiopathology , Cerebrovascular Circulation , Vasodilation , Aging/pathology , Animals , Astrocytes/pathology , Calcium/metabolism , Carbon Dioxide/metabolism , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Male , Mice
19.
Science ; 348(6232): 347-52, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25765066

ABSTRACT

After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier-permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.


Subject(s)
Axons/drug effects , Cicatrix/prevention & control , Epothilones/administration & dosage , Nerve Regeneration/drug effects , Spinal Cord Injuries/drug therapy , Tubulin Modulators/administration & dosage , Animals , Axons/physiology , Cell Movement/drug effects , Cell Polarity/drug effects , Cicatrix/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Meninges/drug effects , Meninges/pathology , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Rats , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
20.
Cell Rep ; 9(4): 1191-201, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456121

ABSTRACT

Clearing techniques have been developed to transparentize mouse brains, thereby preserving 3D structure, but their complexity has limited their use. Here, we show that immunolabeling of axonal tracts followed by optical clearing with solvents (3DISCO) and light-sheet microscopy reveals brain connectivity in mouse embryos and postnatal brains. We show that the Robo3 receptor is selectively expressed by medial habenula axons forming the fasciculus retroflexus (FR) and analyzed the development of this commissural tract in mutants of the Slit/Robo and DCC/Netrin pathways. Netrin-1 and DCC are required to attract FR axons to the midline, but the two mutants exhibit specific and heterogeneous axon guidance defects. Moreover, floor-plate-specific deletion of Slit ligands with a conditional Slit2 allele perturbs not only midline crossing by FR axons but also their anteroposterior distribution. In conclusion, this method represents a unique and powerful imaging tool to study axonal connectivity in mutant mice.


Subject(s)
Axons/metabolism , Brain/metabolism , Imaging, Three-Dimensional/methods , Staining and Labeling , Animals , Biomarkers/metabolism , DCC Receptor , Embryo, Mammalian/metabolism , Membrane Proteins/metabolism , Mice, Knockout , Mutation , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Netrin-1 , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL