Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 109
1.
Nat Commun ; 12(1): 3190, 2021 05 27.
Article En | MEDLINE | ID: mdl-34045465

The hippocampus is essential for spatial and episodic memory but is damaged early in Alzheimer's disease and is very sensitive to hypoxia. Understanding how it regulates its oxygen supply is therefore key for designing interventions to preserve its function. However, studies of neurovascular function in the hippocampus in vivo have been limited by its relative inaccessibility. Here we compared hippocampal and visual cortical neurovascular function in awake mice, using two photon imaging of individual neurons and vessels and measures of regional blood flow and haemoglobin oxygenation. We show that blood flow, blood oxygenation and neurovascular coupling were decreased in the hippocampus compared to neocortex, because of differences in both the vascular network and pericyte and endothelial cell function. Modelling oxygen diffusion indicates that these features of the hippocampal vasculature may restrict oxygen availability and could explain its sensitivity to damage during neurological conditions, including Alzheimer's disease, where the brain's energy supply is decreased.


Hippocampus/blood supply , Microcirculation/physiology , Neocortex/blood supply , Visual Cortex/blood supply , Adenosine Triphosphate/biosynthesis , Alzheimer Disease/physiopathology , Animals , Cell Hypoxia/physiology , Dementia, Vascular/physiopathology , Female , Hippocampus/cytology , Hippocampus/diagnostic imaging , Hippocampus/physiopathology , Humans , Intravital Microscopy , Laser-Doppler Flowmetry , Male , Mice , Microscopy, Fluorescence, Multiphoton , Microvessels/diagnostic imaging , Microvessels/physiology , Models, Animal , Neocortex/cytology , Neocortex/diagnostic imaging , Neocortex/physiopathology , Neurons/metabolism , Neurovascular Coupling/physiology , Oxidative Phosphorylation , Oxygen/analysis , Oxygen/metabolism , Spatial Memory/physiology , Visual Cortex/cytology , Visual Cortex/physiopathology
2.
Fluids Barriers CNS ; 18(1): 14, 2021 Mar 20.
Article En | MEDLINE | ID: mdl-33743764

Central nervous system diseases involving the parenchymal microvessels are frequently associated with a 'microvasculopathy', which includes different levels of neurovascular unit (NVU) dysfunction, including blood-brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches.


Glioma/physiopathology , Neocortex/blood supply , Neocortex/growth & development , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Neural Crest/cytology , Pericytes/physiology , Humans
3.
Cereb Cortex ; 31(4): 2139-2155, 2021 03 05.
Article En | MEDLINE | ID: mdl-33279961

Microglial cells make extensive contacts with neural precursor cells (NPCs) and affiliate with vasculature in the developing cerebral cortex. But how vasculature contributes to cortical histogenesis is not yet fully understood. To better understand functional roles of developing vasculature in the embryonic rat cerebral cortex, we investigated the temporal and spatial relationships between vessels, microglia, and NPCs in the ventricular zone. Our results show that endothelial cells in developing cortical vessels extend numerous fine processes that directly contact mitotic NPCs and microglia; that these processes protrude from vessel walls and are distinct from tip cell processes; and that microglia, NPCs, and vessels are highly interconnected near the ventricle. These findings demonstrate the complex environment in which NPCs are embedded in cortical proliferative zones and suggest that developing vasculature represents a source of signaling with the potential to broadly influence cortical development. In summary, cortical histogenesis arises from the interplay among NPCs, microglia, and developing vasculature. Thus, factors that impinge on any single component have the potential to change the trajectory of cortical development and increase susceptibility for altered neurodevelopmental outcomes.


Cerebral Ventricles/blood supply , Cerebral Ventricles/embryology , Neocortex/blood supply , Neocortex/embryology , Neurogenesis/physiology , Neuroimmunomodulation/physiology , Animals , Cerebral Ventricles/cytology , Embryonic Development/physiology , Female , Microglia/physiology , Neocortex/cytology , Neural Stem Cells/physiology , Pregnancy , Rats
4.
Microvasc Res ; 132: 104059, 2020 11.
Article En | MEDLINE | ID: mdl-32798551

The blood-brain barrier (BBB) maintains the optimal microenvironment for brain function. Tight junctions (TJs) allow endothelial cells to adhere to each other, leading to the formation of a barrier that prevents the penetration of most molecules via transcellular routes. Evidence has indicated that seizure-induced vascular endothelial growth factor (VEGF) type 2 receptor (VEGFR-2) pathway activation weakens TJs, inducing vasodilatation and increasing vascular permeability and subsequent brain injury. The present study focused on investigating the expression levels of VEGF-related (VEGF-A and VEGFR-2) and TJ-related proteins (claudin-5, occludin and ZO-1) in the neocortical microvasculature of patients with drug-resistant temporal lobe epilepsy (TLE). The results obtained from hippocampal sclerosis TLE (HS-TLE) patients were compared with those obtained from patients with TLE secondary to lesions (lesion-TLE) and autopsy samples. The Western blotting and immunofluorescence results showed that VEGF-A and VEGFR-2 protein expression levels were increased in HS-TLE and lesion-TLE patients compared to autopsy group. On the other hand, claudin-5 expression was higher in HS-TLE patients and lesion-TLE patients than autopsies. The expression level of occludin and ZO-1 was decreased in HS-TLE patients. Our study described modifications to the integrity of the BBB that may contribute to the pathogenesis of TLE, in which the VEGF system may play an important role. We demonstrated that the same modifications were present in both HS-TLE and lesion-TLE patients, which suggests that seizures modify these systems and that they are not associated with the establishment of epilepsy.


Blood-Brain Barrier/metabolism , Drug Resistant Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Microvessels/metabolism , Neocortex/blood supply , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adolescent , Adult , Blood-Brain Barrier/pathology , Claudin-5/metabolism , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/pathology , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Female , Humans , Male , Microvessels/pathology , Middle Aged , Occludin/metabolism , Signal Transduction , Tight Junctions/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult , Zonula Occludens-1 Protein/metabolism
5.
Acta Histochem ; 122(5): 151573, 2020 Jul.
Article En | MEDLINE | ID: mdl-32622419

Stroke-induced changes in neuroglia determine the basic conditions for the survival and damage of neurons in the ischemic core. Here, we studied the immunolocalization of glial cell line-derived neurotrophic factor (GDNF), glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba-1), and S-100ß in the rat parietal cortex after constant occlusion of the middle cerebral artery. These cytoplasmic proteins are specific for different glial cell types. They are used as indicators of activated microglia and astrocytes in immunocytochemical studies. The distribution pattern of all markers changed dramatically with time. GFAP- and S-100ß-positive astrocytes were observed in the penumbra zone and marked its boundaries. In days 1-8 after surgery, in the ischemic core, the number of S-100ß-immunoreactive astrocytes decreased, and individual pyramidal cells appeared. S-100ß-expressing pyramidal cells were localized in cortical layers III and V. They were only found in the ischemic core. Their proportion to the total number of cells was 37.3 ± 3.9 %, 22.2 ± 1.2 %, 16.3 ± 2.3 %, and 5.4 ± 0.3 % on days 1, 3, 8, and 14 after surgery. On day 21, no S-100ß-expressing pyramidal cells were observed. The spatial density of GFAP- and S-100ß-positive astrocytes increased in the penumbra region adjacent to the ischemic core and decreased in the penumbral periphery. As a result, the width of the perifocal penumbra zone decreased substantially at later stages of the stroke. In the penumbra, on days 1-3 after ischemic injury, GDNF immunoreactivity was mainly localized in neurons, while later on (days 8-21) it was mainly constrained to astrocyte glia. In intact rats, GDNF-positive neurons were situated in cortical layers II/III and V/VI and made up 52 ± 4.5 % of the total neuron population. Their proportion to the total number of neurons was 29 ± 2.1 %, 13.8 ± 0.6 %, and 3.1 ± 0.2 % on days 1, 8, and 21 after surgery. The number of GDNF-positive astrocytes, on the opposite, increased with time after ischemic injury. Iba-1-reactive microglia was mainly localized to the ischemic core. Microglial cells appeared activated as evidenced by their increased size and decreased number of processes and branching density. The spatial density of microglia reached a peak ​​on day 8 after ischemic injury both in the ischemic core and penumbra. An increase in the number of Iba-1-reactive microglia in the ischemic core correlated with a decrease of the number of GFAP-positive astrocytes. The results are discussed in the context of participation of neuroglia in regulation of various neuroprotective and destructive processes.


Astrocytes/metabolism , Middle Cerebral Artery/metabolism , Neocortex/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Glial Fibrillary Acidic Protein/metabolism , Infarction, Middle Cerebral Artery/metabolism , Male , Microglia/metabolism , Neocortex/blood supply , Rats
7.
J Neurosurg ; 134(5): 1535-1543, 2020 May 22.
Article En | MEDLINE | ID: mdl-32442967

OBJECTIVE: This study aimed to clarify the underlying mechanism of pathognomonic angiogenesis between the temporal muscle and neocortex after indirect bypass for moyamoya disease by shedding light on the role of platelet-derived growth factor receptor-α (PDGFRα) in angiogenesis. METHODS: The gene for PDGFRα was systemically inactivated in adult mice (α-KO mice). The Pdgfra-preserving mice (Flox mice) and α-KO mice were exposed to bilateral common carotid artery stenosis (BCAS) by using microcoils. One week later the animals underwent encephalomyosynangiosis (EMS) on the right side. Cerebral blood flow (CBF) was serially measured using a laser Doppler flowmeter. Histological analysis was performed on the distribution of CD31-positive vessels and collagen deposit at 28 days after BCAS. Reverse transcription polymerase chain reaction (RT-PCR) was performed to assess the expression of collagen mRNA in the skin fibroblasts derived from Flox and α-KO mice. RESULTS: BCAS significantly reduced CBF up to approximately 70% of the control level at 28 days after the onset. There was no significant difference in CBF between Flox and α-KO mice. EMS significantly enhanced the improvement of CBF on the ipsilateral side of Flox mice, but not α-KO mice. EMS significantly induced the development of CD31-positive vessels in both the neocortex and temporal muscle on the ipsilateral side of Flox mice, but not α-KO mice. Deposition of collagen was distinctly observed between them in Flox mice, but not α-KO mice. Expression of mRNA of collagen type 1 alpha 1 (Col1a1) and collagen type 3 alpha 1 (Col3a1) was significantly downregulated in the skin fibroblasts from α-KO mice. CONCLUSIONS: This is the first study that denotes the role of a specific growth factor in angiogenesis after EMS for moyamoya disease by inactivating its gene in mice. The findings strongly suggest that PDGFRα signal may play an important role in developing spontaneous angiogenesis between the temporal muscle and neocortex after EMS in moyamoya disease.


Carotid Stenosis/physiopathology , Cerebral Revascularization/methods , Disease Models, Animal , Moyamoya Disease , Neovascularization, Physiologic/physiology , Receptor, Platelet-Derived Growth Factor alpha/physiology , Animals , Carotid Stenosis/surgery , Cerebrovascular Circulation , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Collagen Type III/biosynthesis , Collagen Type III/genetics , Female , Fibroblasts/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neocortex/blood supply , RNA, Messenger/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/genetics , Temporal Muscle/blood supply
8.
Cell Rep ; 29(5): 1113-1129.e5, 2019 10 29.
Article En | MEDLINE | ID: mdl-31665628

To facilitate efficient oxygen and nutrient delivery, blood vessels in the brain form three-dimensional patterns. However, little is known about how blood vessels develop stereographically in the neocortex and how they control the expansion and differentiation of neural progenitors during neocortical development. We show that highly vascularized and avascular regions are strictly controlled in a spatially and temporally restricted manner and are associated with distinct cell populations. Dividing basal progenitors and oligodendrocyte precursors preferentially contact honeycomb vessels, but dividing apical progenitors are localized in avascular regions without Flt1-positive endothelial cells but directly contact with sprouting neovascular tip cells. Therefore, not all blood vessels are associated equally with neural progenitors. Furthermore, a disruption of normal vascular patterning can induce abnormalities in neural development, whereas the impaired features of neural progenitors influenced angiogenesis patterning. These results indicate that close association between the nervous and vascular systems is essential for neocortex assembly.


Neocortex/cytology , Neocortex/embryology , Neovascularization, Physiologic , Neural Stem Cells/cytology , Animals , Cell Differentiation , Cell Hypoxia , Cell Polarity , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Humans , Integrin beta Chains/metabolism , Male , Mice , Mice, Inbred ICR , Neocortex/blood supply , Neocortex/ultrastructure , Oligodendroglia/cytology , Oligodendroglia/metabolism , Pseudopodia/metabolism , Stem Cell Niche , Time Factors
9.
Neuroimage ; 202: 116067, 2019 11 15.
Article En | MEDLINE | ID: mdl-31394180

Studies of flow-metabolism coupling often presume that microvessel architecture is a surrogate for blood flow. To test this assumption, we introduce an in vivo Dynamic Contrast Optical Coherence Tomography (DyC-OCT) method to quantify layer-resolved microvascular blood flow and volume across the full depth of the mouse neocortex, where the angioarchitecture has been previously described. First, we cross-validate average DyC-OCT cortical flow against conventional Doppler OCT flow. Next, with laminar DyC-OCT, we discover that layer 4 consistently exhibits the highest microvascular blood flow, approximately two-fold higher than the outer cortical layers. While flow differences between layers are well-explained by microvascular volume and density, flow differences between subjects are better explained by transit time. Finally, from layer-resolved tracer enhancement, we also infer that microvascular hematocrit increases in deep cortical layers, consistent with predictions of plasma skimming. Altogether, our results show that while the cortical blood supply derives mainly from the pial surface, laminar hemodynamics ensure that the energetic needs of individual cortical layers are met. The laminar trends reported here provide data that links predictions based on the cortical angioarchitecture to cerebrovascular physiology in vivo.


Cerebrovascular Circulation/physiology , Hemodynamics/physiology , Models, Neurological , Neocortex/blood supply , Neocortex/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Microvessels/anatomy & histology , Microvessels/physiology , Tomography, Optical Coherence
10.
Neuroscience ; 406: 167-175, 2019 May 15.
Article En | MEDLINE | ID: mdl-30867131

Oligodendrocytes (OLGs) differentiate from oligodendrocyte-precursor-cells (OPCs) for myelination in white matter. This differentiation is maintained by cell-cell interactions through trophic factors such as brain-derived-neurotrophic-factor (BDNF). However, differentiation is impaired when white matter injury occurs in a chronic cerebral hypoperfusion model. Thus, we examined the effects of the interaction between astrocyte and oligodendrocyte lineage cells on myelination regarding the mechanism of impairment. A microcoil was applied to the bilateral common carotid arteries in male C57BL/6 mice as an in vivo cerebral chronic hypoperfusion model (BCAS model). A nonlethal concentration of CoCl2 was added to the primary cell culture from the postnatal rat cortex and incubated in vitro. White matter injury progressed in the BCAS model as myelin decreased. The numbers of OPCs and astrocytes increased after the operation, whereas that of OLGs decreased at day 28. BDNF continuously decreased until day 28. Differentiation was disrupted under the stressed conditions in the cell culture, but improved after administration of astrocyte-conditioned medium containing BDNF. Astrocytes with BDNF underwent differentiation, but differentiation was impaired under the stressed conditions due to the reduction of BDNF. We examined S100B regarding the mechanism of impairment. S100B is mainly expressed by mature astrocytes, and has neuroprotective and neurotoxic effects inside and outside of cells. GFAP-positive astrocytes increased in the corpus callosum in the BCAS model, whereas the number of mature astrocytes continued to decrease, resulting in reduced BDNF. The reduction in mature astrocytes due to the discharge of S100B in ischemic conditions caused the reduction in BDNF.


Astrocytes/metabolism , Cell Lineage/physiology , Cerebrovascular Circulation/physiology , Neocortex/metabolism , Oligodendroglia/metabolism , White Matter/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Neocortex/blood supply , Rats , S100 Calcium Binding Protein beta Subunit/metabolism , White Matter/blood supply , White Matter/injuries
11.
Neuroimage ; 197: 792-805, 2019 08 15.
Article En | MEDLINE | ID: mdl-28669910

An amazingly wide range of complex behavior emerges from the cerebral cortex. Much of the information processing that leads to these behaviors is performed in neocortical circuits that span throughout the six layers of the cortex. Maintaining this circuit activity requires substantial quantities of oxygen and energy substrates, which are delivered by the complex yet well-organized and tightly-regulated vascular system. In this review, we provide a detailed characterization of the most relevant anatomical and functional features of the cortical vasculature. This includes a compilation of the available data on laminar variation of vascular density and the topological aspects of the microvascular system. We also review the spatio-temporal dynamics of cortical blood flow regulation and oxygenation, many aspects of which remain poorly understood. Finally, we discuss some of the important implications of vascular density, distribution, oxygenation and blood flow regulation for (laminar) fMRI.


Neocortex/blood supply , Neocortex/physiology , Neurovascular Coupling/physiology , Animals , Functional Neuroimaging/methods , Hemodynamics/physiology , Humans , Magnetic Resonance Imaging/methods
12.
Zh Evol Biokhim Fiziol ; 53(1): 49-54, 2017 01.
Article Ru | MEDLINE | ID: mdl-30695442

Morphological characteristics of the elements of vessel walls in the microvasculature of rat neocortex were studied after perinatal hypoxia and subsequent introduction of salifen, a derivative of GABA, at long-term postnatal periods. It is shown that salifen at a therapeutic dose has a protective effect on the endothelium of blood vessels in the microvasculature of neocortex. It is found that after exposure to hypoxia followed by application of salifen no endothelial hypertrophy, appearance of numerous processes of endotheliocytes in the vascular lumen, and narrowing of the capillary lumen occur. The formation of la- mina densa is almost identical to that in the control. Swelling of perivascular astrocytes and reactive chan- ges of pericytes are also absent. In all layers of the neocortex the density of vessel distribution in the mic- rovasculature as well as their cross-sectional area in adult animals of the control group and after exposure to hypoxia and salifen application were approximately identical. It is shown that changes and restructu- ring of the capillary bed after exposure to hypoxia and application of salifen take place at earlier terms of development whereas by the maturity period stabilization of the structural parameters of the microvas- culature occurs. The protective effect of salifen on the elements of capillary walls determines the clinical effectiveness of its use and serves as a basis for further research in this direction.


Capillaries , Cerebrovascular Circulation , Endothelium, Vascular , Hypoxia , Microcirculation , Neocortex , Prenatal Exposure Delayed Effects , Animals , Capillaries/metabolism , Capillaries/pathology , Capillaries/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/physiopathology , Neocortex/blood supply , Neocortex/metabolism , Neocortex/pathology , Neocortex/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Wistar
13.
Dev Biol ; 420(1): 148-165, 2016 Dec 01.
Article En | MEDLINE | ID: mdl-27671872

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.


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
14.
Age (Dordr) ; 38(4): 273-289, 2016 Aug.
Article En | MEDLINE | ID: mdl-27613724

Strong epidemiological and experimental evidence indicate that both age and hypertension lead to significant functional and structural impairment of the cerebral microcirculation, predisposing to the development of vascular cognitive impairment (VCI) and Alzheimer's disease. Preclinical studies establish a causal link between cognitive decline and microvascular rarefaction in the hippocampus, an area of brain important for learning and memory. Age-related decline in circulating IGF-1 levels results in functional impairment of the cerebral microvessels; however, the mechanistic role of IGF-1 deficiency in impaired hippocampal microvascularization remains elusive. The present study was designed to characterize the additive/synergistic effects of IGF-1 deficiency and hypertension on microvascular density and expression of genes involved in angiogenesis and microvascular regression in the hippocampus. To achieve that goal, we induced hypertension in control and IGF-1 deficient mice (Igf1 f/f  + TBG-Cre-AAV8) by chronic infusion of angiotensin II. We found that circulating IGF-1 deficiency is associated with decreased microvascular density and exacerbates hypertension-induced microvascular rarefaction both in the hippocampus and the neocortex. The anti-angiogenic hippocampal gene expression signature observed in hypertensive IGF-1 deficient mice in the present study provides important clues for subsequent studies to elucidate mechanisms by which hypertension may contribute to the pathogenesis and clinical manifestation of VCI. In conclusion, adult-onset, isolated endocrine IGF-1 deficiency exerts deleterious effects on the cerebral microcirculation, leading to a significant decline in cortical and hippocampal capillarity and exacerbating hypertension-induced cerebromicrovascular rarefaction. The morphological impairment of the cerebral microvasculature induced by IGF-1 deficiency and hypertension reported here, in combination with neurovascular uncoupling, increased blood-brain barrier disruption and neuroinflammation reported in previous studies likely contribute to the pathogenesis of vascular cognitive impairment in elderly hypertensive humans.


Aging/metabolism , Hippocampus/blood supply , Hypertension/complications , Insulin-Like Growth Factor I/deficiency , Microvascular Rarefaction/pathology , Neocortex/blood supply , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Angiotensin II/adverse effects , Angiotensin II/metabolism , Animals , Biomarkers/blood , Blood-Brain Barrier/metabolism , Cognitive Dysfunction/physiopathology , Gene Expression , Humans , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred C57BL , Microvascular Rarefaction/etiology , RNA, Messenger/metabolism
15.
Brain Res ; 1646: 543-550, 2016 09 01.
Article En | MEDLINE | ID: mdl-27369449

Diffusion within the extracellular and perivascular spaces of the brain plays an important role in biological processes, therapeutic delivery, and clearance mechanisms within the central nervous system. Recently, ultrasound has been used to enhance the dispersion of locally administered molecules and particles within the brain, but ultrasound-mediated effects on the brain parenchyma remain poorly understood. We combined an electron microscopy-based ultrastructural analysis with high-resolution tracking of non-adhesive nanoparticles in order to probe changes in the extracellular and perivascular spaces of the brain following a non-destructive pulsed ultrasound regimen known to alter diffusivity in other tissues. Freshly obtained rat brain neocortical slices underwent sham treatment or pulsed, low intensity ultrasound for 5min at 1MHz. Transmission electron microscopy revealed intact cells and blood vessels and evidence of enlarged spaces, particularly adjacent to blood vessels, in ultrasound-treated brain slices. Additionally, ultrasound significantly increased the diffusion rate of 100nm, 200nm, and 500nm nanoparticles that were injected into the brain slices, while 2000nm particles were unaffected. In ultrasound-treated slices, 91.6% of the 100nm particles, 20.7% of the 200nm particles, 13.8% of the 500nm particles, and 0% of the 2000nm particles exhibited diffusive motion. Thus, pulsed ultrasound can have meaningful structural effects on the brain extracellular and perivascular spaces without evidence of tissue disruption.


Extracellular Space/radiation effects , Neocortex/radiation effects , Ultrasonic Waves , Animals , Diffusion , Extracellular Space/metabolism , Nanoparticles/administration & dosage , Neocortex/blood supply , Neocortex/metabolism , Neocortex/ultrastructure , Rats , Rats, Sprague-Dawley
16.
Dev Cell ; 36(6): 624-38, 2016 Mar 21.
Article En | MEDLINE | ID: mdl-27003936

The neocortex contains glutamatergic excitatory neurons and γ-aminobutyric acid (GABA)ergic inhibitory interneurons. Extensive studies have revealed substantial insights into excitatory neuron production. However, our knowledge of the generation of GABAergic interneurons remains limited. Here we show that periventricular blood vessels selectively influence neocortical interneuron progenitor behavior and neurogenesis. Distinct from those in the dorsal telencephalon, radial glial progenitors (RGPs) in the ventral telencephalon responsible for producing neocortical interneurons progressively grow radial glial fibers anchored to periventricular vessels. This progenitor-vessel association is robust and actively maintained as RGPs undergo interkinetic nuclear migration and divide at the ventricular zone surface. Disruption of this association by selective removal of INTEGRIN ß1 in RGPs leads to a decrease in progenitor division, a loss of PARVALBUMIN and SOMATOSTATIN-expressing interneurons, and defective synaptic inhibition in the neocortex. These results highlight a prominent interaction between RGPs and periventricular vessels important for proper production and function of neocortical interneurons.


Interneurons/cytology , Neocortex/blood supply , Neocortex/embryology , Neural Stem Cells/cytology , Telencephalon/blood supply , Telencephalon/embryology , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gestational Age , Green Fluorescent Proteins/metabolism , Integrin beta1/metabolism , Interneurons/metabolism , Median Eminence/blood supply , Median Eminence/embryology , Median Eminence/metabolism , Mice , Mice, Transgenic , Neocortex/metabolism , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Parvalbumins/metabolism , Pregnancy , Preoptic Area/blood supply , Preoptic Area/embryology , Preoptic Area/metabolism , Recombinant Proteins/metabolism , Somatostatin/metabolism , Telencephalon/metabolism
17.
Acta Histochem ; 118(2): 80-9, 2016 Mar.
Article En | MEDLINE | ID: mdl-26643215

Acetylation of nucleosome histones results in relaxation of DNA and its availability for the transcriptional regulators, and is generally associated with the enhancement of gene expression. Although it is well known that activation of a variety of pro-adaptive genes represents a key event in the development of brain hypoxic/ischemic tolerance, the role of epigenetic mechanisms, in particular histone acetylation, in this process is still unexplored. The aim of the present study was to investigate changes in acetylation of histones in vulnerable brain neurons using original well-standardized model of hypobaric hypoxia and preconditioning-induced tolerance of the brain. Using quantitative immunohistochemistry and Western blot, effects of severe injurious hypobaric hypoxia (SH, 180mm Hg, 3h) and neuroprotective preconditioning mode (three episodes of 360mm Hg for 2h spaced at 24h) on the levels of the acetylated proteins and acetylated H3 Lys24 (H3K24ac) in the neocortex and hippocampus of rats were studied. SH caused global repression of the acetylation processes in the neocortex (layers II-III, V) and hippocampus (CA1, CA3) by 3-24h, and this effect was prevented by the preconditioning. Moreover, hypoxic preconditioning remarkably increased the acetylation of H3K24 in response to SH in the brain areas examined. The preconditioning hypoxia without subsequent SH also stimulated acetylation processes in the neocortex and hippocampus. The moderately enhanced expression of the acetylated proteins in the preconditioned rats was maintained for 24h, whereas acetylation of H3K24 was intense but transient, peaked at 3h. The novel data obtained in the present study indicate that large activation of the acetylation processes, in particular acetylation of histones might be essential for the development of brain hypoxic tolerance.


Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Brain Ischemia/metabolism , CA1 Region, Hippocampal/blood supply , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/blood supply , CA3 Region, Hippocampal/metabolism , Cell Hypoxia , Male , Neocortex/blood supply , Neocortex/metabolism , Rats, Wistar
18.
J Cereb Blood Flow Metab ; 35(9): 1411-5, 2015 Sep.
Article En | MEDLINE | ID: mdl-26126870

Ca(2+)-dependent pathways in neurons and astrocyte endfeet initiate changes in arteriole diameter to regulate local brain blood flow. Whether there exists a threshold of synaptic activity in which arteriole diameter is controlled independent of astrocyte endfeet Ca(2+) remains unclear. We used two-photon fluorescence microscopy to examine synaptically evoked synthetic or genetic Ca(2+) indicator signals around penetrating arterioles in acute slices of the rat neocortex. We discovered a threshold below which vasodilation occurred in the absence of endfeet Ca(2+) signals but with consistent neuronal Ca(2+) transients, suggesting endfoot Ca(2+) is not necessary for activity-dependent vasodilation under subtle degrees of brain activation.


Astrocytes/metabolism , Calcium Signaling/physiology , Cerebrovascular Circulation/physiology , Neocortex , Synapses/metabolism , Vasodilation/physiology , Animals , Arterioles/physiology , Astrocytes/cytology , Male , Neocortex/blood supply , Neocortex/cytology , Neocortex/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
19.
Zh Evol Biokhim Fiziol ; 51(5): 377-82, 2015.
Article Ru | MEDLINE | ID: mdl-26856078

For the last time a particular interest of investigators has been attracted to the period of early newborn state when active process of adaptation of the organism to new life conditions occur defining its increased sensitivity to the effect of unfavorable environmental factors. An important place among these processes belongs to formation of homeostasis mechanisms and, primarily, the barrier mechanisms. The purpose of the present study was to investigate reactions of the hematoencephalic barrier (HEB) to action of perinatal normobaric hypoxia (a model of incomplete human pregnancy). Using light and electron microscopy, our investigation showed that after action of hypoxia all wall elements of neocortex capillaries revealed structural alterations which may cause impairment of microcirculation and increased permeability of HEB. It is established that differentiation of the basal membrane of a capillary wall takes place during the early perinatal period and the indicator of its differentiation is the formation of its plates - laminae rara et densa. After action of hypoxia, besides a delay of formation of the basal membrane, a vesicular type of its degeneration occurs. Key words: perinatal hypoxia, hematoencephalic barrier, capillary, endothelial cells, basal membrane.


Asphyxia Neonatorum/pathology , Blood-Brain Barrier/ultrastructure , Hypoxia/pathology , Animals , Capillaries/ultrastructure , Endothelium, Vascular/ultrastructure , Neocortex/blood supply , Rats
20.
Microcirculation ; 21(7): 664-76, 2014 Oct.
Article En | MEDLINE | ID: mdl-24813724

OBJECTIVES: HIV-1 infection of the CNS is associated with impairment of CBF and neurocognitive function, and accelerated signs of aging. As normal aging is associated with rarefaction of the cerebral vasculature, we set out to examine chronic viral effects on the cerebral vasculature. METHODS: DOX-inducible HIV-1 Tat-tg and WT control mice were used. Animals were treated with DOX for three weeks or five to seven months. Cerebral vessel density and capillary segment length were determined from quantitative image analyses of sectioned cortical tissue. In addition, movement of red blood cells in individual capillaries was imaged in vivo using multiphoton microscopy, to determine RBCV and flux. RESULTS: Mean RBCV was not different between Tat-tg mice and age-matched WT controls. However, cortical capillaries from Tat-tg mice showed a significant loss of RBCV heterogeneity and increased RBCF that was attributed to a marked decrease in total cortical capillary length (35-40%) compared to WT mice. CONCLUSIONS: Cerebrovascular rarefaction is accelerated in HIV-1 Tat-transgenic mice, and this is associated with alterations in red cell blood velocity. These changes may have relevance to the pathogenesis of HIV-associated neurocognitive disorders in an aging HIV-positive population.


Blood Flow Velocity , Genes, tat , HIV-1/genetics , Neocortex/blood supply , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , Astrocytes/metabolism , Capillaries/pathology , Doxycycline/pharmacology , Erythrocyte Indices , Hemodynamics , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Neovascularization, Physiologic/drug effects , Pyramidal Cells/pathology , Recombinant Fusion Proteins/toxicity , Up-Regulation/drug effects , tat Gene Products, Human Immunodeficiency Virus/genetics
...