Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Neuron ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38897208

ABSTRACT

Microglia are brain-resident macrophages that contribute to central nervous system (CNS) development, maturation, and preservation. Here, we examine the consequences of permanent microglial deficiencies on brain aging using the Csf1rΔFIRE/ΔFIRE mouse model. In juvenile Csf1rΔFIRE/ΔFIRE mice, we show that microglia are dispensable for the transcriptomic maturation of other brain cell types. By contrast, with advancing age, pathologies accumulate in Csf1rΔFIRE/ΔFIRE brains, macroglia become increasingly dysregulated, and white matter integrity declines, mimicking many pathological features of human CSF1R-related leukoencephalopathy. The thalamus is particularly vulnerable to neuropathological changes in the absence of microglia, with atrophy, neuron loss, vascular alterations, macroglial dysregulation, and severe tissue calcification. We show that populating Csf1rΔFIRE/ΔFIRE brains with wild-type microglia protects against many of these pathological changes. Together with the accompanying study by Chadarevian and colleagues1, our results indicate that the lifelong absence of microglia results in an age-related neurodegenerative condition that can be counteracted via transplantation of healthy microglia.

2.
Brain Pathol ; 33(6): e13189, 2023 11.
Article in English | MEDLINE | ID: mdl-37505935

ABSTRACT

Calcification of the cerebral microvessels in the basal ganglia in the absence of systemic calcium and phosphate imbalance is a hallmark of primary familial brain calcification (PFBC), a rare neurodegenerative disorder. Mutation in genes encoding for sodium-dependent phosphate transporter 2 (SLC20A2), xenotropic and polytropic retrovirus receptor 1 (XPR1), platelet-derived growth factor B (PDGFB), platelet-derived growth factor receptor beta (PDGFRB), myogenesis regulating glycosidase (MYORG), and junctional adhesion molecule 2 (JAM2) are known to cause PFBC. Loss-of-function mutations in XPR1, the only known inorganic phosphate exporter in metazoans, causing dominantly inherited PFBC was first reported in 2015 but until now no studies in the brain have addressed whether loss of one functional allele leads to pathological alterations in mice, a commonly used organism to model human diseases. Here we show that mice heterozygous for Xpr1 (Xpr1WT/lacZ ) present with reduced inorganic phosphate levels in the cerebrospinal fluid and age- and sex-dependent growth of vascular calcifications in the thalamus. Vascular calcifications are surrounded by vascular basement membrane and are located at arterioles in the smooth muscle layer. Similar to previously characterized PFBC mouse models, vascular calcifications in Xpr1WT/lacZ mice contain bone matrix proteins and are surrounded by reactive astrocytes and microglia. However, microglial activation is not confined to calcified vessels but shows a widespread presence. In addition to vascular calcifications, we observed vessel tortuosity and transmission electron microscopy analysis revealed microangiopathy-endothelial swelling, phenotypic alterations in vascular smooth muscle cells, and thickening of the basement membrane.


Subject(s)
Brain Diseases , Neurodegenerative Diseases , Vascular Calcification , Humans , Animals , Mice , Brain Diseases/pathology , Phosphates/metabolism , Brain/pathology , Xenotropic and Polytropic Retrovirus Receptor , Vascular Calcification/metabolism , Vascular Calcification/pathology , Neurodegenerative Diseases/pathology , Mutation , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism
3.
Nat Commun ; 14(1): 3345, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37291094

ABSTRACT

Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds, and in rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and in ex vivo human brain slices, although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial-specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. We apply this approach to Hevin knockout mice, where AAV-X1-mediated ectopic expression of the synaptogenic protein Sparcl1/Hevin in brain endothelial cells rescued synaptic deficits.


Subject(s)
Endothelial Cells , Rodentia , Mice , Rats , Animals , Endothelial Cells/metabolism , Rodentia/genetics , Macaca mulatta/genetics , Brain/metabolism , Tropism/genetics , Mice, Knockout , Dependovirus/metabolism , Genetic Vectors/genetics , Transduction, Genetic , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/genetics
4.
Life Sci ; 321: 121593, 2023 May 15.
Article in English | MEDLINE | ID: mdl-36934970

ABSTRACT

AIMS: Neurovascular-glymphatic dysfunction plays an important role in Alzheimer's disease and has been analysed mainly in relation to amyloid-beta (Aß) pathology. Here, we aim to investigate the neurovascular alterations and mapping of aquaporin 4 (AQP4) distribution and dislocation associated with tau and Aß. MATERIALS AND METHODS: Perfusion, susceptibility weighted imaging and structural magnetic resonance imaging (MRI) were performed in the pR5 mouse model of 4-repeat tau and the arcAß mouse model of amyloidosis. Immunofluorescence staining was performed using antibodies against AQP4, vessel, astroglia, microglia, phospho-tau and Aß in brain tissue slices from pR5, arcAß and non-transgenic mice. KEY FINDINGS: pR5 mice showed regional atrophy, preserved cerebral blood flow, and reduced cerebral vessel density compared to non-transgenic mice, while arcAß mice showed cerebral microbleeds and reduced cerebral vessel density. AQP4 dislocation and peri-tau enrichment in the hippocampus and increased AQP4 levels in the cortex and hippocampus were detected in pR5 mice compared to non-transgenic mice. In comparison, cortical AQP4 dislocation and cortical/hippocampal peri-plaque increases were observed in arcAß mice. Increased expression of reactive astrocytes were detected around the tau inclusions in pR5 mice and Aß plaques in arcAß mice. SIGNIFICANCE: We demonstrated the neurovascular alterations, microgliosis, astrogliosis and increased AQP4 regional expression in pR5 tau and arcAß mice. We observed a divergent region-specific AQP4 dislocation and association with phospho-tau and Aß pathologies.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Aquaporin 4 , tau Proteins , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aquaporin 4/genetics , Aquaporin 4/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , Plaque, Amyloid/pathology , tau Proteins/metabolism
5.
bioRxiv ; 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36711773

ABSTRACT

Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds and rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and ex vivo human brain slices although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. Vasculature-secreted Hevin (a synaptogenic protein) rescued synaptic deficits in a mouse model.

6.
J Cereb Blood Flow Metab ; 43(5): 763-777, 2023 05.
Article in English | MEDLINE | ID: mdl-36545806

ABSTRACT

Pericytes are the mural cells of the microvascular network that are in close contact with underlying endothelial cells. Endothelial-secreted PDGFB leads to recruitment of pericytes to the vessel wall, but this is disrupted in Pdgfbret/ret mice when the PDGFB retention motif is deleted. This results in severely reduced pericyte coverage on blood vessels. In this study, we investigated vascular abnormalities and hemodynamics in Pdgfbret/ret mice throughout the cerebrovascular network and in different cortical layers by in vivo two-photon microscopy. We confirmed that Pdgfbret/ret mice are severely deficient in pericytes throughout the vascular network, with enlarged brain blood vessels and a reduced number of vessel branches. Red blood cell velocity, linear density, and tube hematocrit were reduced in Pdgfbret/ret mice, which may impair oxygen delivery to the tissue. We also measured intravascular PO2 and found that concentrations were higher in cortical Layer 2/3 in Pdgfbret/ret mice, indicative of reduced blood oxygen extraction. Finally, we found that Pdgfbret/ret mice had a reduced capacity for vasodilation in response to an acetazolamide challenge during functional MRI imaging. Taken together, these results suggest that severe pericyte deficiency can lead to vascular abnormalities and altered cerebral blood flow, reminiscent of pathologies such as arteriovenous malformations.


Subject(s)
Endothelial Cells , Pericytes , Mice , Animals , Proto-Oncogene Proteins c-sis/metabolism , Pericytes/metabolism , Disease Models, Animal , Becaplermin/metabolism , Hemodynamics , Oxygen/metabolism
7.
Front Cell Dev Biol ; 10: 849469, 2022.
Article in English | MEDLINE | ID: mdl-35450291

ABSTRACT

Electron microscopy is the primary approach to study ultrastructural features of the cerebrovasculature. However, 2D snapshots of a vascular bed capture only a small fraction of its complexity. Recent efforts to synaptically map neuronal circuitry using volume electron microscopy have also sampled the brain microvasculature in 3D. Here, we perform a meta-analysis of 7 data sets spanning different species and brain regions, including two data sets from the MICrONS consortium that have made efforts to segment vasculature in addition to all parenchymal cell types in mouse visual cortex. Exploration of these data have revealed rich information for detailed investigation of the cerebrovasculature. Neurovascular unit cell types (including, but not limited to, endothelial cells, mural cells, perivascular fibroblasts, microglia, and astrocytes) could be discerned across broad microvascular zones. Image contrast was sufficient to identify subcellular details, including endothelial junctions, caveolae, peg-and-socket interactions, mitochondria, Golgi cisternae, microvilli and other cellular protrusions of potential significance to vascular signaling. Additionally, non-cellular structures including the basement membrane and perivascular spaces were visible and could be traced between arterio-venous zones along the vascular wall. These explorations revealed structural features that may be important for vascular functions, such as blood-brain barrier integrity, blood flow control, brain clearance, and bioenergetics. They also identified limitations where accuracy and consistency of segmentation could be further honed by future efforts. The purpose of this article is to introduce these valuable community resources within the framework of cerebrovascular research. We do so by providing an assessment of their vascular contents, identifying features of significance for further study, and discussing next step ideas for refining vascular segmentation and analysis.

8.
Front Aging Neurosci ; 14: 848495, 2022.
Article in English | MEDLINE | ID: mdl-35309892

ABSTRACT

Vascular calcifications are characterized by the ectopic deposition of calcium and phosphate in the vascular lumen or wall. They are a common finding in computed tomography scans or during autopsy and are often directly related to a pathological condition. While the pathogenesis and functional consequences of vascular calcifications have been intensively studied in some peripheral organs, vascular calcification, and its pathogenesis in the central nervous system is poorly characterized and understood. Here, we review the occurrence of vessel calcifications in the brain in the context of aging and various brain diseases. We discuss the pathomechanism of brain vascular calcification in primary familial brain calcification as an example of brain vessel calcification. A particular focus is the response of microglia to the vessel calcification in the brain and their role in the clearance of calcifications.

9.
Handb Exp Pharmacol ; 273: 33-57, 2022.
Article in English | MEDLINE | ID: mdl-33582883

ABSTRACT

The neurovascular unit (NVU) encompasses all brain cells and underlines that neurons, glia and brain vasculature are in intimate physical and functional association. Brain function is dependent on blood flow and local increases in blood flow in response to neural activity - functional hyperaemia takes place at the NVU. Although this is a vital function of the NVU, many studies have demonstrated that the NVU also performs other tasks. Blood vessels in the brain, which are composed of multiple cell types, are essential for correct brain development. They constitute the niche for brain stem cells, sense the environment and communicate changes to neural tissue, and control the immune quiescence of the CNS. In this brief chapter we will discuss new insights into the biology of NVU, which have further revealed the heterogeneity and complexity of the vascular tree and its neurovascular associations.


Subject(s)
Brain , Neurons , Blood-Brain Barrier/metabolism , Humans , Neuroglia , Neurons/metabolism
10.
World Neurosurg ; 157: 143-151, 2022 01.
Article in English | MEDLINE | ID: mdl-34687935

ABSTRACT

BACKGROUND: The mechanisms of brain arteriovenous malformation (bAVM) development, formation, and progress are still poorly understood. By gaining more knowledge about the molecular signature of bAVM in relation to hemorrhage, we might be able to find biomarkers associated with this serious complication, which can function as a goal for further research and can be a potential target for gene therapy. AIMS: To provide a comprehensive overview of the molecular signature of bAVM-related hemorrhage We conducted a systematic review, following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, of articles published in Embase, Medline, Cochrane central, Scopus, and Chinese databases (CNKI, Wanfang). SUMMARY OF REVIEW: Our search identified 3944 articles, of which 3108 remained after removal of duplicates. After title, abstract, and full-text screening, 31 articles were included for analysis. The results show an overview of molecular characteristics. Several genetic polymorphisms are identified that increase the risk of bAVM rupture by increasing the expression of certain inflammatory cytokines (interleukin [IL]-6, IL-17A, IL-1ß, and tumor necrosis factor-α), NOTCH pathways, matrix metalloproteinase-9, and vascular endothelial growth factor-α. CONCLUSIONS: Several molecular factors are associated with the risk of bAVM-related hemorrhage. These factors are associated with increased inflammation on the cellular level and changes in the endothelium leading to instability of the vessel wall. Further investigation of these biomarkers regarding hemorrhage rates, together with their relationship with noninvasive diagnostic methods, should be a goal of future studies to improve the patient specific risk estimation and future treatment options.


Subject(s)
Arteriovenous Fistula/complications , Arteriovenous Fistula/genetics , Cerebral Hemorrhage/genetics , Intracranial Arteriovenous Malformations/complications , Intracranial Arteriovenous Malformations/genetics , Humans , Polymorphism, Genetic
11.
J Cereb Blood Flow Metab ; 42(2): 264-279, 2022 02.
Article in English | MEDLINE | ID: mdl-34689641

ABSTRACT

Platelet-derived growth factor B (PDGFB) released from endothelial cells is indispensable for pericyte recruitment during angiogenesis in embryonic and postnatal organ growth. Constitutive genetic loss-of-function of PDGFB leads to pericyte hypoplasia and the formation of a sparse, dilated and venous-shifted brain microvasculature with dysfunctional blood-brain barrier (BBB) in mice, as well as the formation of microvascular calcification in both mice and humans. Endothelial PDGFB is also expressed in the adult quiescent microvasculature, but here its importance is unknown. We show that deletion of Pdgfb in endothelial cells in 2-months-old mice causes a slowly progressing pericyte loss leading, at 12-18 months of age, to ≈50% decrease in endothelial:pericyte cell ratio, ≈60% decrease in pericyte longitudinal capillary coverage and >70% decrease in pericyte marker expression. Similar to constitutive loss of Pdgfb, this correlates with increased BBB permeability. However, in contrast to the constitutive loss of Pdgfb, adult-induced loss does not lead to vessel dilation, impaired arterio-venous zonation or the formation of microvascular calcifications. We conclude that PDFGB expression in quiescent adult microvascular brain endothelium is critical for the maintenance of pericyte coverage and normal BBB function, but that microvessel dilation, rarefaction, arterio-venous skewing and calcification reflect developmental roles of PDGFB.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelium, Vascular/metabolism , Lymphokines/metabolism , Pericytes/metabolism , Platelet-Derived Growth Factor/metabolism , Vascular Calcification/metabolism , Animals , Blood-Brain Barrier/pathology , Endothelium, Vascular/pathology , Gene Expression Regulation , Lymphokines/genetics , Mice , Mice, Knockout , Pericytes/pathology , Platelet-Derived Growth Factor/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology
12.
Elife ; 102021 07 06.
Article in English | MEDLINE | ID: mdl-34227466

ABSTRACT

Pericytes have been implicated in various neuropathologies, yet little is known about their function and signaling pathways in health. Here, we characterized calcium dynamics of cortical mural cells in anesthetized or awake Pdgfrb-CreERT2;Rosa26< LSL-GCaMP6s > mice and in acute brain slices. Smooth muscle cells (SMCs) and ensheathing pericytes (EPs), also named as terminal vascular SMCs, revealed similar calcium dynamics in vivo. In contrast, calcium signals in capillary pericytes (CPs) were irregular, higher in frequency, and occurred in cellular microdomains. In the absence of the vessel constricting agent U46619 in acute slices, SMCs and EPs revealed only sparse calcium signals, whereas CPs retained their spontaneous calcium activity. Interestingly, chemogenetic activation of neurons in vivo and acute elevations of extracellular potassium in brain slices strongly decreased calcium activity in CPs. We propose that neuronal activation and an extracellular increase in potassium suppress calcium activity in CPs, likely mediated by Kir2.2 and KATP channels.


Subject(s)
Brain/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Animals , Brain/pathology , Capillaries/metabolism , Female , Male , Mice , Muscle, Smooth, Vascular/diagnostic imaging , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Pericytes/cytology , Pericytes/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Vasoconstriction , Veins/metabolism
13.
Fluids Barriers CNS ; 18(1): 34, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34321020

ABSTRACT

BACKGROUND: Genetic variation in a population has an influence on the manifestation of monogenic as well as multifactorial disorders, with the underlying genetic contribution dependent on several interacting variants. Common laboratory mouse strains used for modelling human disease lack the genetic variability of the human population. Therefore, outcomes of rodent studies show limited relevance to human disease. The functionality of brain vasculature is an important modifier of brain diseases. Importantly, the restrictive interface between blood and brain-the blood-brain barrier (BBB) serves as a major obstacle for the drug delivery into the central nervous system (CNS). Using genetically diverse mouse strains, we aimed to investigate the phenotypic and transcriptomic variation of the healthy BBB in different inbred mouse strains. METHODS: We investigated the heterogeneity of brain vasculature in recently wild-derived mouse strains (CAST/EiJ, WSB/EiJ, PWK/PhJ) and long-inbred mouse strains (129S1/SvImJ, A/J, C57BL/6J, DBA/2J, NOD/ShiLtJ) using different phenotypic arms. We used immunohistochemistry and confocal laser microscopy followed by quantitative image analysis to determine vascular density and pericyte coverage in two brain regions-cortex and hippocampus. Using a low molecular weight fluorescence tracer, sodium fluorescein and spectrophotometry analysis, we assessed BBB permeability in young and aged mice of selected strains. For further phenotypic characterization of endothelial cells in inbred mouse strains, we performed bulk RNA sequencing of sorted endothelial cells isolated from cortex and hippocampus. RESULTS: Cortical vessel density and pericyte coverage did not differ among the investigated strains, except in the cortex, where PWK/PhJ showed lower vessel density compared to NOD/ShiLtJ, and a higher pericyte coverage than DBA/2J. The vascular density in the hippocampus differed among analyzed strains but not the pericyte coverage. The staining patterns of endothelial arteriovenous zonation markers were similar in different strains. BBB permeability to a small fluorescent tracer, sodium fluorescein, was also similar in different strains, except in the hippocampus where the CAST/EiJ showed higher permeability than NOD/ShiLtJ. Transcriptomic analysis of endothelial cells revealed that sex of the animal was a major determinant of gene expression differences. In addition, the expression level of several genes implicated in endothelial function and BBB biology differed between wild-derived and long-inbred mouse strains. In aged mice of three investigated strains (DBA/2J, A/J, C57BL/6J) vascular density and pericyte coverage did not change-expect for DBA/2J, whereas vascular permeability to sodium fluorescein increased in all three strains. CONCLUSIONS: Our analysis shows that although there were no major differences in parenchymal vascular morphology and paracellular BBB permeability for small molecular weight tracer between investigated mouse strains or sexes, transcriptomic differences of brain endothelial cells point to variation in gene expression of the intact BBB. These baseline variances might be confounding factors in pathological conditions that may lead to a differential functional outcome dependent on the sex or genetic polymorphism.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability/physiology , Cerebral Cortex/metabolism , Genetic Variation/physiology , Hippocampus/metabolism , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Female , Fluorescein/administration & dosage , Fluorescein/metabolism , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/metabolism , Genetic Variation/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Species Specificity
14.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33653955

ABSTRACT

Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood-brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret ), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti-ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret ;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.


Subject(s)
Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Homeostasis/immunology , Leukocytes/immunology , Pericytes/immunology , Animals , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Homeostasis/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes/pathology , Mice , Mice, Transgenic , Pericytes/pathology , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
15.
Sci Adv ; 7(9)2021 02.
Article in English | MEDLINE | ID: mdl-33637522

ABSTRACT

Microglia participate in central nervous system (CNS) development and homeostasis and are often implicated in modulating disease processes. However, less is known about the role of microglia in the biology of the neurovascular unit (NVU). In particular, data are scant on whether microglia are involved in CNS vascular pathology. In this study, we use a mouse model of primary familial brain calcification, Pdgfbret/ret , to investigate the role of microglia in calcification of the NVU. We report that microglia enclosing vessel calcifications, coined calcification-associated microglia, display a distinct activation phenotype. Pharmacological ablation of microglia with the CSF1R inhibitor PLX5622 leads to aggravated vessel calcification. Mechanistically, we show that microglia require functional TREM2 for controlling vascular calcification. Our results demonstrate that microglial activity in the setting of pathological vascular calcification is beneficial. In addition, we identify a previously unrecognized function of microglia in halting the expansion of vascular calcification.

16.
Cell ; 184(4): 858-860, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606985

ABSTRACT

Recent findings of an active neuroimmune exchange at brain border regions have challenged the concept of the immune-privileged central nervous system. The study by Rustenhoven et al. in this issue of Cell shows that dural sinuses serve as a conduit for brain-derived antigens to interact with the immune system, allowing in situ immune surveillance.


Subject(s)
Brain , Cranial Sinuses , Humans
17.
Neuro Oncol ; 23(12): 2095-2106, 2021 12 01.
Article in English | MEDLINE | ID: mdl-33560373

ABSTRACT

BACKGROUND: Brain tumors, whether primary or secondary, have limited therapeutic options despite advances in understanding driver gene mutations and heterogeneity within tumor cells. The cellular and molecular composition of brain tumor stroma, an important modifier of tumor growth, has been less investigated to date. Only few studies have focused on the vasculature of human brain tumors despite the fact that the blood-brain barrier (BBB) represents the major obstacle for efficient drug delivery. METHODS: In this study, we employed RNA sequencing to characterize transcriptional alterations of endothelial cells (EC) isolated from primary and secondary human brain tumors. We used an immunoprecipitation approach to enrich for EC from normal brain, glioblastoma (GBM), and lung cancer brain metastasis (BM). RESULTS: Analysis of the endothelial transcriptome showed deregulation of genes implicated in cell proliferation, angiogenesis, and deposition of extracellular matrix (ECM) in the vasculature of GBM and BM. Deregulation of genes defining the BBB dysfunction module was found in both tumor types. We identified deregulated expression of genes in vessel-associated fibroblasts in GBM. CONCLUSION: We characterize alterations in BBB genes in GBM and BM vasculature and identify proteins that might be exploited for developing drug delivery platforms. In addition, our analysis on vessel-associated fibroblasts in GBM shows that the cellular composition of brain tumor stroma merits further investigation.


Subject(s)
Brain Neoplasms , Glioblastoma , Blood-Brain Barrier , Brain Neoplasms/genetics , Endothelial Cells , Glioblastoma/genetics , Humans , Transcriptome
18.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33375813

ABSTRACT

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Subject(s)
Blood-Brain Barrier/metabolism , Pericytes/cytology , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/pathology , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Lymphokines/deficiency , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Pericytes/metabolism , Pericytes/pathology , Platelet-Derived Growth Factor/deficiency , Platelet-Derived Growth Factor/genetics , Single-Cell Analysis , Transcriptome
19.
MAGMA ; 33(6): 769-781, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32468149

ABSTRACT

OBJECTIVE: Brain calcifications are associated with several neurodegenerative diseases. Here, we describe the occurrence of intracranial calcifications as a new phenotype in transgenic P301L mice overexpressing four repeat tau, a model of human tauopathy. MATERIALS AND METHODS: Thirty-six P301L mice (Thy1.2) and ten age-matched non-transgenic littermates of different ages were assessed. Gradient echo data were acquired in vivo and ex vivo at 7 T and 9.4 T for susceptibility-weighted imaging (SWI) and phase imaging. In addition, ex vivo micro-computed tomography (µCT) was performed. Histochemistry and immunohistochemistry were used to investigate the nature of the imaging lesions. RESULTS: SW images revealed regional hypointensities in the hippocampus, cortex, caudate nucleus, and thalamus of P301L mice, which in corresponding phase images indicated diamagnetic lesions. Concomitantly, µCT detected hyperdense lesions, though fewer lesions were observed compared to MRI. Diamagnetic susceptibility lesions in the hippocampus increased with age. The immunochemical staining of brain sections revealed osteocalcin-positive deposits. Furthermore, intra-neuronal and vessel-associated osteocalcin-containing nodules co-localized with phosphorylated-tau (AT8 and AT100) in the hippocampus, while vascular osteocalcin-containing nodules were detected in the thalamus in the absence of phosphorylated-tau deposition. DISCUSSION: SWI and phase imaging sensitively detected intracranial calcifications in the P301L mouse model of human tauopathy.


Subject(s)
Tauopathies , tau Proteins , Animals , Disease Models, Animal , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Tauopathies/diagnostic imaging , X-Ray Microtomography
20.
World Neurosurg ; 139: e807-e811, 2020 07.
Article in English | MEDLINE | ID: mdl-32360923

ABSTRACT

BACKGROUND: Case fatality and poor outcome rates are different between brain arteriovenous malformation-associated intracerebral hemorrhage (bAVM-ICH) and spontaneous intracerebral hemorrhage (SICH). These outcome rates, however, have never been compared in patients who need neurosurgical evacuation of the intracerebral hemorrhage (ICH). OBJECTIVE: To compare the short- and long-term functional outcome between surgically treated patients with bAVM-ICH and SICH. METHODS: We collected data from surgically treated ICH patients at the Department of Neurosurgery, University Hospital Zurich, from January 2015 to July 2018. We performed logistic regression analysis to compare the functional outcome between groups, adjusting for demographics, admission characteristics, and stroke risk factors. RESULTS: A total of 26 bAVM-ICH and 115 SICH patients were included in the final analysis. Patients with bAVM-ICH were younger and less likely to have hypertension without significant differences in ICH volume, hematoma location, intraventricular hemorrhage, and other stroke risk factors. A significantly better functional outcome rate was seen in bAVM-ICH patients at short- and long-term follow-up. These differences remained significant after adjusting for confounders. CONCLUSIONS: patients with a bAVM who need surgical evacuation of an ICH have a more favorable outcome than surgically treated patients with spontaneous ICH, even after correction for confounding factors, such as younger age and less premorbid hypertension.


Subject(s)
Arteriovenous Fistula/complications , Arteriovenous Fistula/surgery , Cerebral Hemorrhage/surgery , Intracranial Arteriovenous Malformations/complications , Intracranial Arteriovenous Malformations/surgery , Treatment Outcome , Adult , Aged , Cerebral Hemorrhage/etiology , Female , Humans , Male , Middle Aged , Neurosurgical Procedures , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...