Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Mar 6.
Published in final edited form as: Methods Mol Biol. 2022;2492:3–24. doi: 10.1007/978-1-0716-2289-6_1

Cells of the Blood-brain Barrier: an Overview of the Neurovascular Unit in Health and Disease

HL McConnell 1,#, A Mishra 1,2
PMCID: PMC9987262  NIHMSID: NIHMS1831314  PMID: 35733036

Abstract

The brain is endowed with highly specialized vasculature that is both structurally and functionally unique compared to vasculature supplying peripheral organs. The blood-brain barrier (BBB) of the cerebral vasculature prevents extravasation of blood products into the brain to protect neural tissue and maintain a homeostatic environment. The BBB also functions as part of a neurovascular unit (NVU), which is comprised of neurons, astrocytes, and microglia in addition to the specialized endothelial cells, mural cells, and the basement membrane of the BBB. Through coordinated intercellular signaling, these cells function as a dynamic unit to tightly regulate brain blood flow, vascular function, neuroimmune responses, and waste clearance. In this chapter, we review the functions of individual NVU components, describe neurovascular coupling as a classic example of NVU function, and discuss archetypal NVU pathophysiology during disease.

Keywords: Neurovascular unit, blood-brain barrier, cerebral blood flow, astrogliosis, neurovascular coupling

Introduction

Vasculature in the central nervous system (CNS) is structurally and functionally unique compared to vasculature that supplies the peripheral organs. To protect neural tissue, which is particularly sensitive to changes in the composition of its interstitial fluid, the cerebral vasculature has evolved to tightly regulate the entry and exit of compounds between the blood and the brain. This blood-brain barrier (BBB) is comprised of several different cell types and encompasses a physical barrier as well as selectively regulated transport mechanisms. Highly specialized endothelial cells line the lumenal surface of blood vessels and abut an ablumenal basement membrane that contains embedded mural cells—pericytes or vascular smooth muscle cells (VSMCs). Astrocytes project endfeet processes that completely ensheathe these components, while their fine processes contact neuronal synapses in the neuropil. Motile microglia are also present within this biological interface. Functionally, these components interact to regulate vascular permeability, neuroimmune responses, cerebral blood flow (CBF), and to maintain a homeostatic CNS environment.

With an evolving understanding of the dynamic signaling interplay that occurs between the BBB components and neurons to support neuronal homeostasis and information processing, the BBB is now often considered in the context of a functional neurovascular unit (NVU) (1). The NVU is a fascinating and clinically relevant disease target. Neurovascular dysfunction is present in numerous neurological diseases, yet whether it is a cause or an effect of a particular disease state is currently not well understood (2). The cells of the CNS are specified and differentiated during early development. Thereafter, with few exceptions, these cells neither turn over nor regenerate effectively. This cell longevity renders the CNS susceptible to factors from the periphery that can disrupt cellular function and survival. Furthermore, as a highly energy-dependent organ with minimal energy stores, the brain relies on a near-constant supply of oxygen, glucose, and other nutrients to drive adenosine triphosphate (ATP) production and maintain metabolic homeostasis. Indeed, no cortical brain cell is located further than 25 μm from a capillary (3). Thus, both from a cell survival and metabolic standpoint, the NVU is critical for CNS health and NVU dysfunction is poised to exacerbate or even initiate neurological disease. Herein, we describe the function of NVU components and the role of the NVU in health and disease.

Components of The Neurovascular Unit

NVU components exist along a three-dimensional vascular network of pial and penetrating arterioles, capillaries, venules, and veins. This vascular network densely permeates all brain tissue to ensure adequate nutrient delivery (1, 4) and clearance of metabolic waste products (5, 6). To maintain CNS homeostasis and meet the fluctuating metabolic demands of the brain tissue, NVU components have functional properties that vary by brain region and vascular zone. Although the NVU is present throughout the vascular tree, we focus largely on the capillary NVU as this is where nutrient exchange occurs and the strongest BBB properties are manifest.

We describe non-neuronal components of the NVU radially, beginning with endothelial cells and progressing outward through the layers of basement membranes (noncellular), mural cells, astrocytes, and finally microglia.

Endothelial Cells

A continuous monolayer of specialized endothelial cells lines the blood vessels of the brain. Unlike the endothelial cells lining the peripheral vasculature, brain vascular endothelial cells are completely confluent and interconnected circumferentially by transmembrane tight junction proteins that prevent the passive transfer of cells and molecules between the blood and the brain based on size, surface electrical charge, and lipid solubility (Figure 1). These tight junctions create a strict seal throughout vascular lumenal surfaces giving rise to in vivo transendothelial electrical resistances of up to ~1800 Ω•cm2 and preventing even the flow of water or small ions across the endothelium (7).

Figure 1.

Figure 1.

Vascular distributions of occludin and laminin blood-brain barrier proteins. Double immunofluorescence confocal microscopy performed with anti-occludin (blue) and anti-laminin (magenta) antibodies on formalin-fixed 50-μm-thick rat brain sections. (A) Arteriolar cross section showing occludin-positive tight junctions between endothelial cells arranged circumferentially around the vascular lumen. Laminin-containing basement membrane circumscribes the occludin staining and delineates both the endothelial-secreted vascular basement membrane (arrowhead) and the astrocyte-secreted parenchymal basement membrane layer (arrow). Vascular smooth muscle cells occupy the space between the layers of laminin (not labeled). (B) Surface view of an arteriole shows occludin-positive tight junctions outlining the contiguous and elongated endothelial cells. Laminin staining reveals basement membrane coverage along the entire vessel. (C) View of a capillary branch point shows occludin tight junction staining between adjacent endothelial cells. The laminin-positive vascular and parenchymal basement membranes are fused along most regions along a capillary and indistinguishable except in regions encasing pericytes (arrow). Merged images are shown on the right.

Besides this physical barrier, brain vascular endothelial cells maintain a dynamic regulatory system that facilitates bidirectional transport of substances between the blood and the brain. The transport functions of these cells are mediated and maintained by polarized expression of various lumenal and ablumenal membrane receptors and transporters, such as HCO3- exchangers and Na+/K+-ATPase (8, 9). As opposed to peripheral vascular systems where substances in the blood are more or less indiscriminately moved across the endothelial lining into the tissue via transcytosis, these pathways are suppressed in brain endothelial cells, reducing their non-specific transport capacity (10, 11). Thus, depending on the chemical entity, transport of substances across the BBB must be actively regulated. This occurs via several mechanisms, including paracellular diffusion (12), carrier and receptor-mediated transcytosis (13, 14), and adsorptive-mediated transcytosis (Figure 2) (15, 16). These specialized properties of brain endothelial cells are reflected in their unique gene expression patterns, a finding highlighted in recent transcriptomic studies (1719). For example, expression of Mfsd2a, the transcytosis-suppressing protein responsible for lipid transport regulation, is high in endothelial cells of the cerebral microvasculature but very low in lung and heart endothelial cells (18, 19). Although this regulation effectively maintains the delicate ionic milieu and extracellular homeostasis of neural tissue, it also creates a pharmacological bottleneck for CNS drug delivery (15, 20).

Figure 2.

Figure 2.

Specialized brain vascular endothelial cells regulate transport at the blood-brain barrier. A contiguous monolayer of vascular endothelial cells lines the brain vasculature. These cells are interconnected by tight junctions, creating a physical barrier with a remarkably high transendothelial electrical resistance. Transport across the blood-brain barrier is mediated by three energy-independent diffusion mechanisms and three energy-dependent transport mechanisms. The energy-independent mechanisms include transcellular diffusion, paracellular diffusion, and passive diffusion. The energy-dependent mechanisms encompass receptor-mediated transcytosis, carrier-mediated transport, and adsorptive-mediated transport. Transport is shown only unidirectionally for simplicity. Created with BioRender.com.

Brain vascular endothelial cells are phenotypically heterogeneous and have numerous location-specific functional roles that vary with both their position in the vascular hierarchy and location in the brain (18, 21, 22). For example, the circumventricular organs and hypothalamic regions contain specialized endothelial cells that make their vasculature more permeable than the rest of the brain (21, 22). In addition to their barrier functions, brain vascular endothelial cells dynamically modify permeability in response to inflammatory and immunological stimuli (19) or in a circadian rhythmic fashion (2325). Further, endothelial cells also help mediate neurovascular coupling (NVC) (2628) and regulate the spread of vascular responses via endothelial gap junctions, which allow propagation of signals along the vascular tree to mediate changes in regional CBF (Figure 4) (29, 30). The increased mitochondrial load of brain vascular endothelial cells reflects their active role in NVU regulation, but also makes them more sensitive to changes in oxidative stress (31).

Figure 4.

Figure 4.

Neurovascular coupling (NVC) is the phenomenon by which active neurons increase local blood flow to satisfy their energy demands. During synaptic activity (circled), post-synaptic neurons (magenta) can directly release vasoactive substances onto vessels to change the luminal diameter (I). Synaptic activity can also stimulate astrocytes (purple) to release vasoactive substances via their endfeet (II). Vascular endothelial cells (red) may also respond to signals from astrocytes, and perhaps neurons, to release vasoactive substances onto vessels (III). Regulation of lumenal diameter at arterioles occurs via signaling from neurons and endothelial cells to vascular smooth muscle cells (pink), while astrocytes can mediate NVC by signaling to capillary pericytes (green) and modulate the response of arteriolar smooth muscle cells. Further, endothelial cells, pericytes, and vascular smooth muscle cells can also propagate the spread of vascular responses via gap junctions to mediate regional changes in CBF. Through this coordinated intercellular signaling, NVC components function as a dynamic functional unit to tightly regulate brain blood flow. Inset shows positioning of astrocytes, pericytes, basement membrane (brown), and endothelial cells within a capillary cross-section. Created with BioRender.com.

Basement Membrane

The cerebrovascular endothelial tube is ensheathed by a non-cellular basement membrane comprised of five major proteins—collagens, laminins, nidogens, perlecan, and agrin—arranged in a compact lattice meshwork. This vascular basement membrane merges with a parenchymal basement membrane in areas along the vasculature lacking mural cell coverage, particularly in capillary regions not covered by pericytes (Figure 1). Vascular basement membrane proteins are secreted largely by endothelial cells while parenchymal basement membrane components are secreted by astrocytes via their vascular endfeet (32). Bidirectional signaling between the cells of the NVU—endothelial cells, pericytes, and astrocytes—and the basement membrane dynamically regulates BBB permeability. For example, pericyte recruitment to endothelial tubes during development is an important stimulus that helps generate the basement membrane matrix (33), but absence of the basement membrane protein laminin can, in turn, reduce pericyte coverage of the vasculature (34).

Structurally, basement membrane proteins form a sheet-like extracellular matrix that stabilizes the BBB, anchors cells, facilitates signal transduction, and enhances vascular barrier properties (35). The composition and thickness of the basement membrane are dynamic, vary regionally throughout the brain, and can affect cellular interactions and receptor activation states (35, 36). Functionally, the basement membrane contributes to BBB development, endothelial cell tight junction formation, and maintenance of transendothelial electrical resistance (35). It also helps recruit and establish the polarity of astrocyte endfeet, including anchoring of aquaporin 4 to the vessel-facing cell membrane of the endfoot, functions that are likely mediated via interactions of laminin with its receptor dystroglycan in astrocyte endfeet (34). However, as astrocytes are an important source of the parenchymal basement membrane components, astrocyte dysfunction can also result in a weakened basement membrane (37), underscoring the importance of bidirectional crosstalk between BBB components.

Mural Cells: Pericytes

Mural cells called pericytes enwrap capillaries in all vascular beds. Capillaries in the CNS are associated with the highest pericyte coverage in the body, exhibiting a pericyte to endothelial cell ratio of 1:4 (38, 39) compared to, for example, a ratio of 1:10 or more in skeletal muscle (40, 41). This suggests an important role of pericytes in the cerebral circulation. Indeed, brain pericytes play critical roles during both embryogenesis and adulthood by establishing and maintaining BBB integrity (42), orchestrating vascular development (43), and regulating capillary blood flow (44). During brain development, pericytes migrate to vessels and, in concert with endothelial cells and other NVU components, contribute to vascular pruning and angiogenesis (43, 45). A balance of these processes effectively shapes and organizes the cerebral vasculature to ensure an optimal angioarchitecture that facilitates nutrient delivery to brain tissue. Like the endothelial cells and basement membrane, pericytes tune the polarization of astrocyte endfeet along the capillary wall (46, 47). Embedded between the endothelial tube and astrocyte endfeet, pericytes are often described as having a “bump on a log” morphology (Figure 3) (48). This description accurately describes the cell body but may not adequately convey the extensive coverage of the vasculature by pericyte processes, which can contact up to 90% of brain vasculature in mice (49). Although pericytes are fixed in place and do not migrate after development under physiologically healthy conditions, they are structurally dynamic and can extend their processes and remodel to compensate for loss of nearby pericytes or endothelial cell coverage in areas along the vasculature (50). In adult brains, pericytes exhibit contractile responses to vasoactive compounds and therefore control capillary diameter (44, 51, 52). However, not all pericytes are contractile (5355) and they may be relatively less contractile than the VSMCs on larger vessels. It has been proposed that pericytes may maintain basal CBF equilibrium at the capillary level while arterial VSMCs are responsible for larger, more rapid CBF fluctuations (50).

Figure 3.

Figure 3.

Astrocyte ensheathment of the cerebral vasculature. (A) A capillary network in a mouse retina showing the relationship of astrocytes to pericytes. Astrocytes are labeled with an anti-glial fibrillary acidic protein (GFAP) antibody (green) in a retina whole mount from an NG2-dsRed mouse, wherein the red fluorescent protein dsRed is expressed in NG2-positive pericytes. The vascular lumen is labeled with an anti-mouse secondary (blue) that labels natural mouse serum IgGs present in blood vessels (retina post-fixed following live dissection without perfusion). Astrocyte endfeet encapsulate the capillary wall and contact all pericytes. (B-F) Astrocyte endfeet around a cortical capillary with the surrounding neuropil. (B) An astrocyte labeled with anti-GFAP antibody (red). (C) The same astrocyte expressing virally induced cytosolic green fluorescent protein (GFP) under the truncated GFAP promoter, GfaABC1D (green). (D) The surrounding neuropil labeled with the neuronal marker, microtubule-associated protein 2 (MAP2; magenta). (E) Nuclei are labeled with DAPI (blue). (F) Merged image showing GFAP, GFP, MAP2, and DAPI. Though GFAP labeling shows only the cytoskeleton of the astrocyte, including the endfoot process, GFP expression reveals its extensive fine processes permeating the neuropil (where it contacts synapses) and its complete ensheathment of the capillary via its endfoot.

Pericytes display broad heterogeneity in function, gene expression, and appearance. They are morphologically variable along the vascular tree, showing different structures and vessel coverage at capillary, pre-capillary, and post-capillary vascular zones. Therefore, depending on their location and morphology, pericytes may exert different functions in their regulation of the BBB and CBF at different levels of the vascular tree (48, 56). Pericytes located at branch points between arterioles and capillaries may act as ‘precapillary sphincters’ to direct flow into or away from a region (51, 57). There is also strong evidence that pericytes on pre-capillary arterioles and capillaries closer to arterioles participate in NVC (44) and regulate CBF (50, 58). Gap-junctional connections between neighboring pericytes and between pericytes and endothelial cells also help propagate signals along the vascular tree (30), such that a contractile response initiated in one region quickly spreads up- and down-stream to increase blood flow through that region (59, 60). Pericytes in mid-capillary branches may be more important for basal CBF regulation (61, 62) and maintenance of BBB integrity and stability in concert with endothelial cells (48, 56, 63). Very little is known about the pericytes on cerebral venules, but they are hypothesized to regulate immune cell trafficking, as they do in peripheral vascular beds (64, 65).

Mural Cells: Vascular Smooth Muscle Cells

VSMCs are a distinct contractile mural cell type from pericytes (66). They are embedded in the arterial wall where they form several concentric layers around these larger vessels. They have important roles in maintaining and generating basal vascular tone, vascular reactivity, and autoregulation (67, 68). VSMCs express a variety of ion channels, are electrically excitable, and display robust calcium signals and consequent contractile responses; hence, they are traditionally thought to be the principal site of functional hyperemia in the brain (1, 67). Furthermore, VSMCs are gap junction-coupled to each other (69) and also to endothelial cells lining the vasculature, which allows evoked vasomotor responses to propagate along vessels (70) over long distances as calcium waves (71). The polarity and magnitude of these calcium signals determine the degree of evoked vasoconstriction or dilation. Fluctuations between relaxation and contraction states in cerebral VSMCs gives rise to vasomotion, which is postulated to be a mechanism of perivascular drainage, which clears waste from the brain interstitium (72).

Astrocytes

Astrocytes are the most numerous glial cell type in the CNS with a complex, polarized morphology. On the one hand, they have vascular endfeet processes, which almost completely encapsulate the vasculature of the brain, including the endothelial tube and surrounding mural cells (Figure 3) (73). On the other hand, they have numerous fine, ramified processes extending from a stellate cell body which permeate the neuropil (Figure 3). Like neurons, astrocytes are heterogeneous in structure, function, and distribution throughout the brain. Traditionally, astrocytes were classified into two groups: protoplasmic astrocytes found in the well-vascularized grey matter and fibrous astrocytes found in the less vascular white matter. In both grey and white matter areas, astrocytes physically separate synapses or nodes of Ranvier, contact neurons, and cover the vasculature (74). More recent work has revealed considerable heterogeneity even within grey matter astrocytes, with pronounced regional or even sub-regional differences (7579), some of which are regulated by signals from surrounding NVU components, including neurons (80) and endothelial cells (81, 82). This heterogeneity manifests as variations in astrocyte functions, their interactions with neurons (83, 84), and their response to injuries (85).

Astrocytes play a multifaceted role in maintaining CNS homeostasis. They spatially tile the brain parenchyma and are interconnected by gap junction proteins to form a functional syncytium (86), which allows them to signal over long ranges via calcium wave propagation (87). Astrocytes also express high levels of the inwardly rectifying potassium channel Kir4.1, which is pronounced in their peri-synaptic processes and vascular endfeet. This allows astrocytes to take up potassium after bouts of neuronal activity and spatially buffer potassium along the syncytium (86). At synapses, astrocytes are active participants in tuning the information transfer between neurons by taking up neurotransmitters at the cleft to spatially and temporally refine synaptic signaling (74).

Astrocyte endfeet processes terminate on and enwrap all capillaries and arterioles in the CNS to form a contiguous gliovascular interface. Astrocytes extend other fine processes to a variety of targets within the CNS, including synapses, nerve cell bodies, and nodes of Ranvier. This polarized morphology positions astrocytes centrally within the NVU to dynamically mediate many regulatory functions, including maintenance of BBB integrity, mechanical and metabolic support, endothelial transport, innate immune responses, and cerebrovascular regulation (88, 89). Via their endfeet processes, astrocytes secrete basement membrane proteins such as laminins (90) to stabilize the BBB. They also signal to pericytes (90) and endothelial cells (91) to maintain the CNS-specific properties of these vascular cells within the BBB. Specialized membrane proteins anchored to astrocyte endfeet processes allow astrocytes to regulate the transport of nutrients and neurotransmitters after they cross the BBB, including glucose and glutamate, which are necessary for healthy brain function (81, 92). Further, selective expression of channels such as Kir4.1 and aquaporin 4 at the endfeet polarize astrocytes and facilitate the directional flow of potassium and water molecules to maintain ion homeostasis and neuropil volume (9395).

Astrocytes serve important roles as sensors of neuronal activity and stabilizers of the extracellular environment. They are also active players in information processing within the brain. In response to synaptic activity, numerous intracellular signaling pathways are activated in astrocytes, the most studied so far being calcium-dependent. Calcium increases can be transient and localized within microdomains, spread through an entire process, spread throughout the astrocyte territory, or even be sustained and propagated across the astrocyte syncytium (87, 96). This activation of astrocytes results in the release of gliotransmitters (at a spatial extent dictated by the calcium signals), which can then modulate activity ranging from anywhere between individual synapses to whole circuits (25, 87, 9799). Given that astrocytes also express virtually all neuromodulator receptors, they have been proposed to be the cellular substrates underlying coordinated long-range neuromodulatory networks (100). With their endfeet on vessels for nutrient uptake and their gap junction-coupled fine neuropil processes, astrocytes also form a crucial metabolic supply network that not only regulates glucose delivery to active regions (101) but may also modulate circuit properties that regulate plasticity (102) and behaviors such as sleep (103).

Within the NVU, astrocytes are key regulators of CBF (104). Astrocytes contribute to the resting tone of arterioles through release of vasoactive agents such as prostaglandin E2 (105) and ATP (106, 107). They can mediate signals from active neurons to the vasculature to mediate NVC. This intercellular signaling pathway – from neurons to astrocytes to the vascular mural cells – is the primary pathway regulating capillary NVC (29, 52, 108), but it appears to be less important in regulating arteriolar NVC, where it is engaged only upon sustained or strong activation (Figure 4) (109). Even so, recruitment of astrocytes results in a threefold enhancement of functional hyperemia in vivo, suggesting an important role for astrocytes in activity-dependent energy supply to the brain (110).

Finally, astrocytes serve an important role in glymphatic waste clearance. During sleep, aquaporin-4 water transport channels on the vessel-facing endfeet processes of astrocytes facilitate the exchange of perivascular cerebrospinal fluid (CSF) with interstitial fluid at capillaries (111). This exchange results in the drainage of CNS waste products into the CSF, which is then removed at CSF clearance sites. Both sleep deprivation (112) and aging (113) result in glymphatic dysfunction by causing mislocalization of aquaporin 4 on astrocyte endfeet, potentially contributing to the accumulation of toxic waste products in the brain.

Microglia

Microglia are yolk sac-derived myeloid cells that seed the brain early in development (114). During this early period, microglia stimulate angiogenesis, increase vascular complexity, and promote endothelial tip cell fusion within capillary beds (115117). Conversely, the vascular network may also provide microglia with a migration ‘highway’ to populate the brain during development (118). Adult microglia are the tissue-resident macrophages of the brain. These highly ramified, motile cells act as phagocytic sentinels, surveilling and probing their environment for any alterations or injury by extending and retracting their processes. Microglia are scattered evenly throughout the brain, in a manner similar to the tiling of astrocytes, and occasionally extend a process directly to the vasculature at locations containing pericytes (73). They are exquisitely sensitive to changes in the neuropil environment or in nearby perivascular homeostasis. Upon sensing insult, microglia take on an ameboid morphology and undergo dynamic transcriptomic and phenotypic changes that are characterized along a spectrum of pro-inflammatory to anti-inflammatory activated states. These activated microglia serve a critical role in phagocytosing debris (119, 120), signaling to peripheral immune cells (120), and regulating reactive astrogliosis (121, 122). Microglia may also modulate the BBB, sometimes enhancing BBB properties and sometimes contributing to BBB breakdown and NVU dysfunction (123125), depending on the type, extent, and chronicity of the injury (126128). As with any other component of the NVU, microglial properties are also dynamically modulated by surrounding NVU cells such as astrocytes and endothelial cells (129). Together, astroglial and microglial activation may optimize innate immune responses in the CNS by engaging in coordinated gliotransmitter and cytokine release and metabolite uptake (130).

Neurovascular Coupling as a Classic Example of Intercellular NVU Signaling

The CNS is dependent on a continuous supply of energy substrates and other nutrients from the blood to maintain healthy function. Signaling within the NVU, particularly between astrocytes and mural cells, regulates the resting tone of CNS vessels to help maintain resting blood flow (105, 107). Subcortical projection neurons originating from regions such as the basal ganglia and the brainstem can endow cerebral vessels with vascular tone, which may occur directly, via astrocytes, or via local interneurons (reviewed in (29)). The brain is also protected from large fluctuations in blood flow due to changes in systemic physiology via a process termed cerebral autoregulation, such that blood flow is maintained relatively constant within a physiological range of blood pressure changes. Although autoregulation is generally mediated by myogenic or endothelial factors, a role for astrocytes in a process akin to autoregulation was recently identified, whereby astrocytes sense pressure changes and release vasoconstrictors onto mural cells to maintain vascular tone (106). Layered upon these processes is NVC, an intercellular signaling mechanism that engages almost all cellular components of the NVU to increase blood supply to regions of increased neuronal activity in a spatially and temporally coordinated manner (Figure 4) (104), and gives rise to a functional hyperemia response in the brain. Neuronal activity and blood flow changes are coupled so tightly that this phenomenon has been exploited by various functional neuroimaging techniques such as blood oxygenation level-dependent functional magnetic resonance imaging (29, 131, 132), positron emissions tomography (133, 134), and the recently developed functional ultrasound (135) to image brain activity in real-time.

The study of pathways regulating NVC has blossomed in the last two decades, revealing complex intercellular interactions at the NVU interface. NVC is initiated by neuronal activity and can be mediated by vasoactive signals released directly from neurons or indirectly via astrocytes, which trigger a change in vascular lumen diameter (Figure 4) (reviewed in (1, 29, 67)). Vasodilatory signals such as nitric oxide (NO), prostaglandins, potassium, and epoxyeicosatrienoic acids released by neurons and/or astrocytes can relax mural cells, thereby dilating blood vessels and increasing CBF. Conversely, vasoconstrictive signals such as ATP and 20-hydroxytetraenoic acids from neurons/astrocytes cause contraction of mural cells, thereby constricting blood vessels and decreasing CBF. Though traditional views held that NVC only involved direct signaling from neurons to the VSMCs to increase CBF at the arteriolar level, recent studies have found significant evidence for a role of pericytes in capillary flow regulation and for astrocytes in mediating signals to capillaries (Figure 5) (52, 108). Indeed, with their fine processes apposed to numerous synapses and their endfeet ensheathing the entire cerebral vasculature (including arterioles and capillaries) astrocytes are ideally positioned to be NVC intermediaries. Current evidence suggests that astrocytes play an obligatory role in capillary NVC (52, 108), while their role in arteriolar NVC may be more modulatory (109, 110, 136). Ultimately, signals from both neurons and astrocytes must alter the contractile state of vascular mural cells—pericytes on capillaries or VSMCs on arterioles—for NVC to successfully produce functional hyperemia. Furthermore, a role for endothelial cells in this intercellular signaling pathway, wherein endothelial cells respond to signals from astrocytes and in turn secrete vasodilators onto VMSCs, has also been discovered (Figure 4) (28). Thus, functional interactions between NVU components are crucial regulators of blood flow and hence energy supply to the brain.

Figure 5.

Figure 5.

Neuronal stimulation evokes dilation of capillaries in the cortex. (A-E) A 300-μm-thick acute rat brain slice preparation showing a cortical capillary and the surrounding neuropil. A recording electrode for measuring field potentials is also visible. Successive images show the response of the capillary to the preconstrictor U46619 (B), electrical stimulation of cortical layer I/II axons (C), recovery from stimulation-evoked response (D) and after washout of U46619 (E). (F) The diameter of the capillary region marked by arrowheads in A-E is plotted over time, demonstrating a strong constriction in response to the thromboxane analog U46619 (200 nM), followed by a brief dilation in response to neuronal stimulation. Washing out U46619 reverses the constricted tone (end of trace). Notably, the changes in capillary diameter occur only in a restricted segment while the neighboring region remains undisturbed (arrow). Such a response is characteristic of pericyte-mediated diameter changes. This response is blocked by inhibitors of action potentials and post-synaptic activity, indicating that it is dependent on neuronal activity (data not shown, see (52) for more details). (G) A field recording of neuronal activity evoked by electrical stimulation of layer 1/II in the same experiment (detected by the recording electrode visible in A). The fiber volley (compound action potentials) and the field excitatory post-synaptic current are clearly visible. A 3 s, 20 Hz stimulus train was applied to induce the capillary response shown in F.

NVU in Disease

Neurovascular dysfunction is a central theme of numerous brain disease states, though whether it is a cause or an effect of a particular disease state is not always well defined. Broadly, neurovascular dysfunction encompasses a range of pathological states that can be attributed to individual NVU components or the unit as a whole. In lieu of a particular disease focus, we instead describe elements of NVU dysfunction that are common to many brain diseases. Critically, NVU dysfunction occurs along a spectrum with variable pathophysiological features depending on the disease state and individual.

As a major function of the NVU is to maintain BBB characteristics and function, a general consequence of NVU dysfunction is often BBB disruption, though this term can reflect distinct and nuanced structural, functional, cellular, and molecular deficits depending on the particular disease state. For example, NVU cellular responses to hypoxia differ from those occurring in neurodegeneration or traumatic injury. On a structural level, BBB disruption reflects a loss of integrity and an increase in permeability due to altered endothelial cell expression of tight junction proteins (137, 138) and transporters (139), enlarged perivascular spaces (140), or even degeneration of NVU components including endothelial cells (141), pericytes (142144), astrocytes (145147), or basement membranes (148). Pathologically impaired waste clearance may also compromise BBB integrity (149). A functional consequence of increased BBB permeability is permissive infiltration of blood products through the vulnerable barrier, including but not limited to peripheral immune cell infiltrates.

Under conditions that deviate from a normal physiological range, astrocytes often become ‘reactive’ and alter their morphologic and transcriptomic properties. Reactive astrogliosis can also be induced following BBB breakdown via signaling from blood-borne proteins such as albumin, fibronectin, and thrombin (2, 150152). Reactive astrocytes are characterized by gain of new functions or loss of existing functions, which together alter the overall functions of the region affected (2, 153). They can also release various cytokines that can alter the properties of nearby neurons, microglia, and vascular cells (154), and perhaps even attract or modulate peripheral immune cells to the rescue of the CNS (155157). The current consensus is that reactive astrogliosis is a context-dependent process engaged to re-establish homeostasis and protect neurons from further damage (122, 158160); however, in some contexts, they acquire detrimental properties and may amplify injury (121, 161, 162). The central positioning of astrocytes in neurovascular signaling also poises them to subtly or dramatically impair energy homeostasis in the CNS due to an “uncoupling” of neuronal activity and vascular responses during both acute and chronic NVU impairment (2, 104). Indeed, CBF alterations are commonly seen in many brain disease states and can manifest as decreased perfusion to deep brain structures, loss of NVC, and impaired vascular reactivity to flow and pressure changes (163167).

Conclusion

The mammalian cerebrovascular system is characterized by structurally and functionally unique vessels endowed with a strong biological barrier that selectively separates blood and brain components. Distinct functional features characterize individual NVU components, though they maintain many intimate functional relationships with one another. Further, cells of the NVU can change both functionally and phenotypically, depending on microenvironmental signaling cues.

The presence of the BBB has important implications for neuronal function during physiology and pathology. It protects the CNS extracellular environment from peripheral signals that could destabilize neuronal function. Further, complex intercellular signals between NVU components allows these cells to sense and respond to changes in regional neuronal activity to regulate CBF and meet the energy demands of neurons. The recent development and application of novel and sophisticated neuroimaging paradigms that allow real-time imaging of these physiological processes offers exciting prospects for delineating the contributions of individual NVU components to regional hemodynamic changes.

Finally, NVU dysfunction can precede or exacerbate CNS disease states. With their multifaceted functional roles in physiological processes, dysfunctional NVU components are poised to adversely influence a broad range of metabolic, hemodynamic, and immunologic regulatory responses in the brain. Compelling evidence so far supports this view; however, considerable research will be required to form a complete picture of the NVU’s role in neurological diseases.

Acknowledgments

The authors thank Dr. Leslie Muldoon, Dr. Sasha Pejerrey, and Adrienne Winston for helpful comments on the manuscript, and gratefully acknowledge Dr. Zhihua Wang for assistance with the immunohistochemistry shown in Figure 1. H.L.M was supported by a Ruth L. Kirschstein National Research Service Award T32HL094294 (NIH). A.M.’s lab is supported by NIH grants R01NS110690 (NINDS), R01DA047237 (NIMH; PI Andrew Adey) and a Development Project grant under P30AG066518-01; Oregon Alzheimer Disease Research Center (NIA; PI Jeffrey Kaye).

References

  • 1.Iadecola C (2017) The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron 96(1):17–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.McConnell HL, Li Z, Woltjer RL, and Mishra A (2019) Astrocyte dysfunction and neurovascular impairment in neurological disorders: Correlation or causation? Neurochem Int 128:70–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lovick TA, Brown LA, and Key BJ (1999) Neurovascular relationships in hippocampal slices: physiological and anatomical studies of mechanisms underlying flow-metabolism coupling in intraparenchymal microvessels. Neuroscience 92(1):47–60. [DOI] [PubMed] [Google Scholar]
  • 4.Blinder P, Tsai PS, Kaufhold JP, Knutsen PM, Suhl H, and Kleinfeld D (2013) The cortical angiome: an interconnected vascular network with noncolumnar patterns of blood flow. Nat Neurosci 16(7):889–897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, et al. (2013) Sleep drives metabolite clearance from the adult brain. Science 342(6156):373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kisler K, Nelson AR, Montagne A, and Zlokovic BV (2017) Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci 18(7):419–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Butt AM, Jones HC, and Abbott NJ (1990) Electrical resistance across the blood-brain barrier in anaesthetized rats: a developmental study. J Physiol 429:47–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Pluimer BR, Colt M, and Zhao Z (2020) G Protein-Coupled Receptors in the Mammalian Blood-Brain Barrier. Front Cell Neurosci 14:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cornford EM, and Hyman S (2005) Localization of brain endothelial luminal and abluminal transporters with immunogold electron microscopy. NeuroRx 2(1):27–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Andreone BJ, Chow BW, Tata A, Lacoste B, Ben-Zvi A, Bullock K, et al. (2017) Blood-Brain Barrier Permeability Is Regulated by Lipid Transport-Dependent Suppression of Caveolae-Mediated Transcytosis. Neuron 94(3):581–594 e585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chow BW, and Gu C (2017) Gradual Suppression of Transcytosis Governs Functional Blood-Retinal Barrier Formation. Neuron 93(6):1325–1333 e1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Keaney J, Walsh DM, O’Malley T, Hudson N, Crosbie DE, Loftus T, et al. (2015) Autoregulated paracellular clearance of amyloid-beta across the blood-brain barrier. Sci Adv 1(8):e1500472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Urayama A, Grubb JH, Sly WS, and Banks WA (2004) Developmentally regulated mannose 6-phosphate receptor-mediated transport of a lysosomal enzyme across the blood-brain barrier. Proc Natl Acad Sci U S A 101(34):12658–12663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Pardridge WM, Triguero D, Yang J, and Cancilla PA (1990) Comparison of in vitro and in vivo models of drug transcytosis through the blood-brain barrier. J Pharmacol Exp Ther 253(2):884–891. [PubMed] [Google Scholar]
  • 15.Lombardo SM, Schneider M, Tureli AE, and Gunday Tureli N (2020) Key for crossing the BBB with nanoparticles: the rational design. Beilstein J Nanotechnol 11:866–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Matsumoto J, Stewart T, Sheng L, Li N, Bullock K, Song N, et al. (2017) Transmission of alpha-synuclein-containing erythrocyte-derived extracellular vesicles across the blood-brain barrier via adsorptive mediated transcytosis: another mechanism for initiation and progression of Parkinson’s disease? Acta Neuropathol Commun 5(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, and Barres BA (2010) The mouse blood-brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS One 5(10):e13741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.He L, Vanlandewijck M, Mae MA, Andrae J, Ando K, Del Gaudio F, et al. (2018) Single-cell RNA sequencing of mouse brain and lung vascular and vessel-associated cell types. Sci Data 5:180160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jambusaria A, Hong Z, Zhang L, Srivastava S, Jana A, Toth PT, et al. (2020) Endothelial heterogeneity across distinct vascular beds during homeostasis and inflammation. Elife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lochhead JJ, Yang J, Ronaldson PT, and Davis TP (2020) Structure, Function, and Regulation of the Blood-Brain Barrier Tight Junction in Central Nervous System Disorders. Front Physiol 11:914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ganong WF (2000) Circumventricular organs: definition and role in the regulation of endocrine and autonomic function. Clin Exp Pharmacol Physiol 27(5–6):422–427. [DOI] [PubMed] [Google Scholar]
  • 22.Rodriguez EM, Blazquez JL, and Guerra M (2010) The design of barriers in the hypothalamus allows the median eminence and the arcuate nucleus to enjoy private milieus: the former opens to the portal blood and the latter to the cerebrospinal fluid. Peptides 31(4):757–776. [DOI] [PubMed] [Google Scholar]
  • 23.Cuddapah VA, Zhang SL, and Sehgal A (2019) Regulation of the Blood-Brain Barrier by Circadian Rhythms and Sleep. Trends Neurosci 42(7):500–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhang SL, Yue Z, Arnold DM, Artiushin G, and Sehgal A (2018) A Circadian Clock in the Blood-Brain Barrier Regulates Xenobiotic Efflux. Cell 173(1):130–139 e110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gomez-Gonzalez B, Hurtado-Alvarado G, Esqueda-Leon E, Santana-Miranda R, Rojas-Zamorano JA, and Velazquez-Moctezuma J (2013) REM sleep loss and recovery regulates blood-brain barrier function. Curr Neurovasc Res 10(3):197–207. [DOI] [PubMed] [Google Scholar]
  • 26.Atochin DN, and Huang PL (2011) Role of endothelial nitric oxide in cerebrovascular regulation. Curr Pharm Biotechnol 12(9):1334–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hogan-Cann AD, Lu P, and Anderson CM (2019) Endothelial NMDA receptors mediate activity-dependent brain hemodynamic responses in mice. Proc Natl Acad Sci U S A 116(21):10229–10231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Stobart JL, Lu L, Anderson HD, Mori H, and Anderson CM (2013) Astrocyte-induced cortical vasodilation is mediated by D-serine and endothelial nitric oxide synthase. Proc Natl Acad Sci U S A 110(8):3149–3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Howarth C, Mishra A, and Hall CN (2021) More than just summed neuronal activity: how multiple cell types shape the BOLD response. Philos Trans R Soc Lond B Biol Sci 376(1815):20190630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kovacs-Oller T, Ivanova E, Bianchimano P, and Sagdullaev BT (2019) Dynamic connectivity maps of pericytes and endothelial cells mediate neurovascular coupling in health and disease. bioRxiv:830398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Davidson SM, and Duchen MR (2007) Endothelial mitochondria: contributing to vascular function and disease. Circ Res 100(8):1128–1141. [DOI] [PubMed] [Google Scholar]
  • 32.Castro Dias M, Mapunda JA, Vladymyrov M, and Engelhardt B (2019) Structure and Junctional Complexes of Endothelial, Epithelial and Glial Brain Barriers. Int J Mol Sci 20(21). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stratman AN, Malotte KM, Mahan RD, Davis MJ, and Davis GE (2009) Pericyte recruitment during vasculogenic tube assembly stimulates endothelial basement membrane matrix formation. Blood 114(24):5091–5101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Menezes MJ, McClenahan FK, Leiton CV, Aranmolate A, Shan X, and Colognato H (2014) The extracellular matrix protein laminin alpha2 regulates the maturation and function of the blood-brain barrier. J Neurosci 34(46):15260–15280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Xu L, Nirwane A, and Yao Y (2019) Basement membrane and blood-brain barrier. Stroke Vasc Neurol 4(2):78–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ceafalan LC, Fertig TE, Gheorghe TC, Hinescu ME, Popescu BO, Pahnke J, et al. (2019) Age-related ultrastructural changes of the basement membrane in the mouse blood-brain barrier. J Cell Mol Med 23(2):819–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Robel S, Mori T, Zoubaa S, Schlegel J, Sirko S, Faissner A, et al. (2009) Conditional deletion of beta1-integrin in astroglia causes partial reactive gliosis. Glia 57(15):1630–1647. [DOI] [PubMed] [Google Scholar]
  • 38.Bonkowski D, Katyshev V, Balabanov RD, Borisov A, and Dore-Duffy P (2011) The CNS microvascular pericyte: pericyte-astrocyte crosstalk in the regulation of tissue survival. Fluids Barriers CNS 8(1):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Smyth LCD, Rustenhoven J, Scotter EL, Schweder P, Faull RLM, Park TIH, et al. (2018) Markers for human brain pericytes and smooth muscle cells. J Chem Neuroanat 92:48–60. [DOI] [PubMed] [Google Scholar]
  • 40.Armulik A, Genove G, and Betsholtz C (2011) Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 21(2):193–215. [DOI] [PubMed] [Google Scholar]
  • 41.Geevarghese A, and Herman IM (2014) Pericyte-endothelial crosstalk: implications and opportunities for advanced cellular therapies. Transl Res 163(4):296–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Daneman R, Zhou L, Kebede AA, and Barres BA (2010) Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 468(7323):562–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gaengel K, Genove G, Armulik A, and Betsholtz C (2009) Endothelial-mural cell signaling in vascular development and angiogenesis. Arterioscler Thromb Vasc Biol 29(5):630–638. [DOI] [PubMed] [Google Scholar]
  • 44.Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, et al. (2014) Capillary pericytes regulate cerebral blood flow in health and disease. Nature 508(7494):55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Korn C, and Augustin HG (2015) Mechanisms of Vessel Pruning and Regression. Dev Cell 34(1):5–17. [DOI] [PubMed] [Google Scholar]
  • 46.Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet C, et al. (2010) Pericytes regulate the blood-brain barrier. Nature 468(7323):557–561. [DOI] [PubMed] [Google Scholar]
  • 47.Gundersen GA, Vindedal GF, Skare O, and Nagelhus EA (2014) Evidence that pericytes regulate aquaporin-4 polarization in mouse cortical astrocytes. Brain Struct Funct 219(6):2181–2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Attwell D, Mishra A, Hall CN, O’Farrell FM, and Dalkara T (2016) What is a pericyte? J Cereb Blood Flow Metab 36(2):451–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Grant RI, Hartmann DA, Underly RG, Berthiaume AA, Bhat NR, and Shih AY (2019) Organizational hierarchy and structural diversity of microvascular pericytes in adult mouse cortex. J Cereb Blood Flow Metab 39(3):411–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Berthiaume AA, Hartmann DA, Majesky MW, Bhat NR, and Shih AY (2018) Pericyte Structural Remodeling in Cerebrovascular Health and Homeostasis. Front Aging Neurosci 10:210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gonzales AL, Klug NR, Moshkforoush A, Lee JC, Lee FK, Shui B, et al. (2020) Contractile pericytes determine the direction of blood flow at capillary junctions. Proc Natl Acad Sci U S A 117(43):27022–27033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mishra A, Reynolds JP, Chen Y, Gourine AV, Rusakov DA, and Attwell D (2016) Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles. Nat Neurosci 19(12):1619–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Alarcon-Martinez L, Yilmaz-Ozcan S, Yemisci M, Schallek J, Kilic K, Villafranca-Baughman D, et al. (2019) Retinal ischemia induces alpha-SMA-mediated capillary pericyte contraction coincident with perivascular glycogen depletion. Acta Neuropathol Commun 7(1):134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Bandopadhyay R, Orte C, Lawrenson JG, Reid AR, De Silva S, and Allt G (2001) Contractile proteins in pericytes at the blood-brain and blood-retinal barriers. J Neurocytol 30(1):35–44. [DOI] [PubMed] [Google Scholar]
  • 55.Nehls V, and Drenckhahn D (1991) Heterogeneity of microvascular pericytes for smooth muscle type alpha-actin. J Cell Biol 113(1):147–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Arango-Lievano M, Boussadia B, De Terdonck LDT, Gault C, Fontanaud P, Lafont C, et al. (2018) Topographic Reorganization of Cerebrovascular Mural Cells under Seizure Conditions. Cell Rep 23(4):1045–1059. [DOI] [PubMed] [Google Scholar]
  • 57.Grubb S, Cai C, Hald BO, Khennouf L, Murmu RP, Jensen AGK, et al. (2020) Precapillary sphincters maintain perfusion in the cerebral cortex. Nat Commun 11(1):395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Montagne A, Nikolakopoulou AM, Zhao Z, Sagare AP, Si G, Lazic D, et al. (2018) Pericyte degeneration causes white matter dysfunction in the mouse central nervous system. Nat Med 24(3):326–337. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 59.Cai C, Fordsmann JC, Jensen SH, Gesslein B, Lonstrup M, Hald BO, et al. (2018) Stimulation-induced increases in cerebral blood flow and local capillary vasoconstriction depend on conducted vascular responses. Proc Natl Acad Sci U S A 115(25):E5796–E5804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rungta RL, Chaigneau E, Osmanski BF, and Charpak S (2018) Vascular Compartmentalization of Functional Hyperemia from the Synapse to the Pia. Neuron 99(2):362–375 e364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hartmann DA, Berthiaume A-A, Grant RI, Harrill SA, Noonan T, Costello J, et al. (2020) Brain capillary pericytes exert a substantial but slow influence on blood flow. bioRxiv:2020.2003.2026.008763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Nelson AR, Sagare MA, Wang Y, Kisler K, Zhao Z, and Zlokovic BV (2020) Channelrhodopsin Excitation Contracts Brain Pericytes and Reduces Blood Flow in the Aging Mouse Brain in vivo. Front Aging Neurosci 12:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Profaci CP, Munji RN, Pulido RS, and Daneman R (2020) The blood-brain barrier in health and disease: Important unanswered questions. J Exp Med 217(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Proebstl D, Voisin MB, Woodfin A, Whiteford J, D’Acquisto F, Jones GE, et al. (2012) Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J Exp Med 209(6):1219–1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sims DE, Miller FN, Donald A, and Perricone MA (1990) Ultrastructure of pericytes in early stages of histamine-induced inflammation. J Morphol 206(3):333–342. [DOI] [PubMed] [Google Scholar]
  • 66.Damisah EC, Hill RA, Tong L, Murray KN, and Grutzendler J (2017) A fluoro-Nissl dye identifies pericytes as distinct vascular mural cells during in vivo brain imaging. Nat Neurosci 20(7):1023–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Attwell D, Buchan AM, Charpak S, Lauritzen M, Macvicar BA, and Newman EA (2010) Glial and neuronal control of brain blood flow. Nature 468(7321):232–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Iadecola C, and Nedergaard M (2007) Glial regulation of the cerebral microvasculature. Nat Neurosci 10(11):1369–1376. [DOI] [PubMed] [Google Scholar]
  • 69.Christ GJ, Spray DC, el-Sabban M, Moore LK, and Brink PR (1996) Gap junctions in vascular tissues. Evaluating the role of intercellular communication in the modulation of vasomotor tone. Circ Res 79(4):631–646. [DOI] [PubMed] [Google Scholar]
  • 70.Welsh DG, Tran CHT, Hald BO, and Sancho M (2018) The Conducted Vasomotor Response: Function, Biophysical Basis, and Pharmacological Control. Annu Rev Pharmacol Toxicol 58:391–410. [DOI] [PubMed] [Google Scholar]
  • 71.Seppey D, Sauser R, Koenigsberger M, Beny JL, and Meister JJ (2010) Intercellular calcium waves are associated with the propagation of vasomotion along arterial strips. Am J Physiol Heart Circ Physiol 298(2):H488–496. [DOI] [PubMed] [Google Scholar]
  • 72.Aldea R, Weller RO, Wilcock DM, Carare RO, and Richardson G (2019) Cerebrovascular Smooth Muscle Cells as the Drivers of Intramural Periarterial Drainage of the Brain. Front Aging Neurosci 11:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mathiisen TM, Lehre KP, Danbolt NC, and Ottersen OP (2010) The perivascular astroglial sheath provides a complete covering of the brain microvessels: an electron microscopic 3D reconstruction. Glia 58(9):1094–1103. [DOI] [PubMed] [Google Scholar]
  • 74.Sofroniew MV, and Vinters HV (2010) Astrocytes: biology and pathology. Acta Neuropathol 119(1):7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Batiuk MY, Martirosyan A, Wahis J, de Vin F, Marneffe C, Kusserow C, et al. (2020) Identification of region-specific astrocyte subtypes at single cell resolution. Nat Commun 11(1):1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bayraktar OA, Bartels T, Holmqvist S, Kleshchevnikov V, Martirosyan A, Polioudakis D, et al. (2020) Astrocyte layers in the mammalian cerebral cortex revealed by a single-cell in situ transcriptomic map. Nat Neurosci 23(4):500–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chai H, Diaz-Castro B, Shigetomi E, Monte E, Octeau JC, Yu X, et al. (2017) Neural Circuit-Specialized Astrocytes: Transcriptomic, Proteomic, Morphological, and Functional Evidence. Neuron 95(3):531–549 e539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.John Lin CC, Yu K, Hatcher A, Huang TW, Lee HK, Carlson J, et al. (2017) Identification of diverse astrocyte populations and their malignant analogs. Nat Neurosci 20(3):396–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tsai HH, Li H, Fuentealba LC, Molofsky AV, Taveira-Marques R, Zhuang H, et al. (2012) Regional astrocyte allocation regulates CNS synaptogenesis and repair. Science 337(6092):358–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Farmer WT, Abrahamsson T, Chierzi S, Lui C, Zaelzer C, Jones EV, et al. (2016) Neurons diversify astrocytes in the adult brain through sonic hedgehog signaling. Science 351(6275):849–854. [DOI] [PubMed] [Google Scholar]
  • 81.Lee ML, Martinez-Lozada Z, Krizman EN, and Robinson MB (2017) Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 143(5):489–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mi H, Haeberle H, and Barres BA (2001) Induction of astrocyte differentiation by endothelial cells. J Neurosci 21(5):1538–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hu X, Qin S, Huang X, Yuan Y, Tan Z, Gu Y, et al. (2019) Region-Restrict Astrocytes Exhibit Heterogeneous Susceptibility to Neuronal Reprogramming. Stem Cell Reports 12(2):290–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ziemens D, Oschmann F, Gerkau NJ, and Rose CR (2019) Heterogeneity of Activity-Induced Sodium Transients between Astrocytes of the Mouse Hippocampus and Neocortex: Mechanisms and Consequences. J Neurosci 39(14):2620–2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rusnakova V, Honsa P, Dzamba D, Stahlberg A, Kubista M, and Anderova M (2013) Heterogeneity of astrocytes: from development to injury - single cell gene expression. PLoS One 8(8):e69734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Scemes E, and Spray DC. The Astrocyte Syncytium. Non-Neural Cells in the Nervous System. Elsevier; 2003:165–179. [Google Scholar]
  • 87.Araque A, Carmignoto G, Haydon PG, Oliet SH, Robitaille R, and Volterra A (2014) Gliotransmitters travel in time and space. Neuron 81(4):728–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Boulay AC, Saubamea B, Adam N, Chasseigneaux S, Mazare N, Gilbert A, et al. (2017) Translation in astrocyte distal processes sets molecular heterogeneity at the gliovascular interface. Cell Discov 3:17005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Li L, Acioglu C, Heary RF, and Elkabes S (2020) Role of astroglial toll-like receptors (TLRs) in central nervous system infections, injury and neurodegenerative diseases. Brain Behav Immun. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yao Y, Chen ZL, Norris EH, and Strickland S (2014) Astrocytic laminin regulates pericyte differentiation and maintains blood brain barrier integrity. Nat Commun 5:3413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Garcia CM, Darland DC, Massingham LJ, and D’Amore PA (2004) Endothelial cell-astrocyte interactions and TGF beta are required for induction of blood-neural barrier properties. Brain Res Dev Brain Res 152(1):25–38. [DOI] [PubMed] [Google Scholar]
  • 92.Kacem K, Lacombe P, Seylaz J, and Bonvento G (1998) Structural organization of the perivascular astrocyte endfeet and their relationship with the endothelial glucose transporter: a confocal microscopy study. Glia 23(1):1–10. [PubMed] [Google Scholar]
  • 93.Amiry-Moghaddam M, Williamson A, Palomba M, Eid T, de Lanerolle NC, Nagelhus EA, et al. (2003) Delayed K+ clearance associated with aquaporin-4 mislocalization: phenotypic defects in brains of alpha-syntrophin-null mice. Proc Natl Acad Sci U S A 100(23):13615–13620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ishii M, Horio Y, Tada Y, Hibino H, Inanobe A, Ito M, et al. (1997) Expression and clustered distribution of an inwardly rectifying potassium channel, KAB-2/Kir4.1, on mammalian retinal Muller cell membrane: their regulation by insulin and laminin signals. J Neurosci 17(20):7725–7735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Manley GT, Fujimura M, Ma T, Noshita N, Filiz F, Bollen AW, et al. (2000) Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med 6(2):159–163. [DOI] [PubMed] [Google Scholar]
  • 96.Bazargani N, and Attwell D (2016) Astrocyte calcium signaling: the third wave. Nat Neurosci 19(2):182–189. [DOI] [PubMed] [Google Scholar]
  • 97.Covelo A, and Araque A (2018) Neuronal activity determines distinct gliotransmitter release from a single astrocyte. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mederos S, Sanchez-Puelles C, Esparza J, Valero M, Ponomarenko A, and Perea G (2020) GABAergic signaling to astrocytes in the prefrontal cortex sustains goal-directed behaviors. Nat Neurosci. [DOI] [PubMed] [Google Scholar]
  • 99.Shigetomi E, Jackson-Weaver O, Huckstepp RT, O’Dell TJ, and Khakh BS (2013) TRPA1 channels are regulators of astrocyte basal calcium levels and long-term potentiation via constitutive D-serine release. J Neurosci 33(24):10143–10153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pacholko AG, Wotton CA, and Bekar LK (2020) Astrocytes-The Ultimate Effectors of Long-Range Neuromodulatory Networks? Front Cell Neurosci 14:581075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Rouach N, Koulakoff A, Abudara V, Willecke K, and Giaume C (2008) Astroglial metabolic networks sustain hippocampal synaptic transmission. Science 322(5907):1551–1555. [DOI] [PubMed] [Google Scholar]
  • 102.Murphy-Royal C, Johnston AD, Boyce AKJ, Diaz-Castro B, Institoris A, Peringod G, et al. (2020) Stress gates an astrocytic energy reservoir to impair synaptic plasticity. Nat Commun 11(1):2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Clasadonte J, Scemes E, Wang Z, Boison D, and Haydon PG (2017) Connexin 43-Mediated Astroglial Metabolic Networks Contribute to the Regulation of the Sleep-Wake Cycle. Neuron 95(6):1365–1380 e1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mishra A (2017) Binaural blood flow control by astrocytes: listening to synapses and the vasculature. J Physiol 595(6):1885–1902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rosenegger DG, Tran CH, Wamsteeker Cusulin JI, and Gordon GR (2015) Tonic Local Brain Blood Flow Control by Astrocytes Independent of Phasic Neurovascular Coupling. J Neurosci 35(39):13463–13474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kim KJ, Iddings JA, Stern JE, Blanco VM, Croom D, Kirov SA, et al. (2015) Astrocyte contributions to flow/pressure-evoked parenchymal arteriole vasoconstriction. J Neurosci 35(21):8245–8257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kur J, and Newman EA (2014) Purinergic control of vascular tone in the retina. J Physiol 592(3):491–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Biesecker KR, Srienc AI, Shimoda AM, Agarwal A, Bergles DE, Kofuji P, et al. (2016) Glial Cell Calcium Signaling Mediates Capillary Regulation of Blood Flow in the Retina. J Neurosci 36(36):9435–9445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Institoris A, Rosenegger DG, and Gordon GR (2015) Arteriole dilation to synaptic activation that is sub-threshold to astrocyte endfoot Ca2+ transients. J Cereb Blood Flow Metab 35(9):1411–1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gu X, Chen W, Volkow ND, Koretsky AP, Du C, and Pan Y (2018) Synchronized Astrocytic Ca(2+) Responses in Neurovascular Coupling during Somatosensory Stimulation and for the Resting State. Cell Rep 23(13):3878–3890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. (2012) A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 4(147):147ra111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Liu DX, He X, Wu D, Zhang Q, Yang C, Liang FY, et al. (2017) Continuous theta burst stimulation facilitates the clearance efficiency of the glymphatic pathway in a mouse model of sleep deprivation. Neurosci Lett 653:189–194. [DOI] [PubMed] [Google Scholar]
  • 113.Kress BT, Iliff JJ, Xia M, Wang M, Wei HS, Zeppenfeld D, et al. (2014) Impairment of paravascular clearance pathways in the aging brain. Ann Neurol 76(6):845–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Thion MS, Ginhoux F, and Garel S (2018) Microglia and early brain development: An intimate journey. Science 362(6411):185–189. [DOI] [PubMed] [Google Scholar]
  • 115.Coelho-Santos V, and Shih AY (2020) Postnatal development of cerebrovascular structure and the neurogliovascular unit. Wiley Interdiscip Rev Dev Biol 9(2):e363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Rymo SF, Gerhardt H, Wolfhagen Sand F, Lang R, Uv A, and Betsholtz C (2011) A two-way communication between microglial cells and angiogenic sprouts regulates angiogenesis in aortic ring cultures. PLoS One 6(1):e15846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, et al. (2010) Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood 116(5):829–840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Mondo E, Becker SC, Kautzman AG, Schifferer M, Baer CE, Chen J, et al. (2020) A Developmental Analysis of Juxtavascular Microglia Dynamics and Interactions with the Vasculature. J Neurosci 40(34):6503–6521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Tanaka T, Ueno M, and Yamashita T (2009) Engulfment of axon debris by microglia requires p38 MAPK activity. J Biol Chem 284(32):21626–21636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Fenn AM, Hall JC, Gensel JC, Popovich PG, and Godbout JP (2014) IL-4 signaling drives a unique arginase+/IL-1beta+ microglia phenotype and recruits macrophages to the inflammatory CNS: consequences of age-related deficits in IL-4Ralpha after traumatic spinal cord injury. J Neurosci 34(26):8904–8917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. (2017) Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541(7638):481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Shinozaki Y, Shibata K, Yoshida K, Shigetomi E, Gachet C, Ikenaka K, et al. (2017) Transformation of Astrocytes to a Neuroprotective Phenotype by Microglia via P2Y1 Receptor Downregulation. Cell Rep 19(6):1151–1164. [DOI] [PubMed] [Google Scholar]
  • 123.Haruwaka K, Ikegami A, Tachibana Y, Ohno N, Konishi H, Hashimoto A, et al. (2019) Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat Commun 10(1):5816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Jolivel V, Bicker F, Biname F, Ploen R, Keller S, Gollan R, et al. (2015) Perivascular microglia promote blood vessel disintegration in the ischemic penumbra. Acta Neuropathol 129(2):279–295. [DOI] [PubMed] [Google Scholar]
  • 125.Halder SK, and Milner R (2019) A critical role for microglia in maintaining vascular integrity in the hypoxic spinal cord. Proc Natl Acad Sci U S A 116(51):26029–26037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Boche D, Perry VH, and Nicoll JA (2013) Review: activation patterns of microglia and their identification in the human brain. Neuropathol Appl Neurobiol 39(1):3–18. [DOI] [PubMed] [Google Scholar]
  • 127.Ronaldson PT, and Davis TP (2020) Regulation of blood-brain barrier integrity by microglia in health and disease: A therapeutic opportunity. J Cereb Blood Flow Metab:271678X20951995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Thurgur H, and Pinteaux E (2019) Microglia in the Neurovascular Unit: Blood-Brain Barrier-microglia Interactions After Central Nervous System Disorders. Neuroscience 405:55–67. [DOI] [PubMed] [Google Scholar]
  • 129.Xing C, Li W, Deng W, Ning M, and Lo EH (2018) A potential gliovascular mechanism for microglial activation: differential phenotypic switching of microglia by endothelium versus astrocytes. J Neuroinflammation 15(1):143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Osipova ED, Semyachkina-Glushkovskaya OV, Morgun AV, Pisareva NV, Malinovskaya NA, Boitsova EB, et al. (2018) Gliotransmitters and cytokines in the control of blood-brain barrier permeability. Rev Neurosci 29(5):567–591. [DOI] [PubMed] [Google Scholar]
  • 131.Logothetis NK (2003) The underpinnings of the BOLD functional magnetic resonance imaging signal. J Neurosci 23(10):3963–3971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Raichle ME (1998) Behind the scenes of functional brain imaging: a historical and physiological perspective. Proc Natl Acad Sci U S A 95(3):765–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Fox PT, Mintun MA, Raichle ME, Miezin FM, Allman JM, and Van Essen DC (1986) Mapping human visual cortex with positron emission tomography. Nature 323(6091):806–809. [DOI] [PubMed] [Google Scholar]
  • 134.Frackowiak RS, and Friston KJ (1994) Functional neuroanatomy of the human brain: positron emission tomography--a new neuroanatomical technique. J Anat 184 ( Pt 2):211–225. [PMC free article] [PubMed] [Google Scholar]
  • 135.Mace E, Montaldo G, Osmanski BF, Cohen I, Fink M, and Tanter M (2013) Functional ultrasound imaging of the brain: theory and basic principles. IEEE Trans Ultrason Ferroelectr Freq Control 60(3):492–506. [DOI] [PubMed] [Google Scholar]
  • 136.Schulz K, Sydekum E, Krueppel R, Engelbrecht CJ, Schlegel F, Schroter A, et al. (2012) Simultaneous BOLD fMRI and fiber-optic calcium recording in rat neocortex. Nat Methods 9(6):597–602. [DOI] [PubMed] [Google Scholar]
  • 137.Jiao H, Wang Z, Liu Y, Wang P, and Xue Y (2011) Specific role of tight junction proteins claudin-5, occludin, and ZO-1 of the blood-brain barrier in a focal cerebral ischemic insult. J Mol Neurosci 44(2):130–139. [DOI] [PubMed] [Google Scholar]
  • 138.Yang Y, Estrada EY, Thompson JF, Liu W, and Rosenberg GA (2007) Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J Cereb Blood Flow Metab 27(4):697–709. [DOI] [PubMed] [Google Scholar]
  • 139.Sweeney MD, Sagare AP, and Zlokovic BV (2018) Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 14(3):133–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Li Y, Li M, Yang L, Qin W, Yang S, Yuan J, et al. (2019) The relationship between blood-brain barrier permeability and enlarged perivascular spaces: a cross-sectional study. Clin Interv Aging 14:871–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Krueger M, Bechmann I, Immig K, Reichenbach A, Hartig W, and Michalski D (2015) Blood-brain barrier breakdown involves four distinct stages of vascular damage in various models of experimental focal cerebral ischemia. J Cereb Blood Flow Metab 35(2):292–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Halliday MR, Rege SV, Ma Q, Zhao Z, Miller CA, Winkler EA, et al. (2016) Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. J Cereb Blood Flow Metab 36(1):216–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kisler K, Nelson AR, Rege SV, Ramanathan A, Wang Y, Ahuja A, et al. (2017) Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain. Nat Neurosci 20(3):406–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Watson AN, Berthiaume AA, Faino AV, McDowell KP, Bhat NR, Hartmann DA, et al. (2020) Mild pericyte deficiency is associated with aberrant brain microvascular flow in aged PDGFRbeta(+/−) mice. J Cereb Blood Flow Metab 40(12):2387–2400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Chen A, Akinyemi RO, Hase Y, Firbank MJ, Ndung’u MN, Foster V, et al. (2016) Frontal white matter hyperintensities, clasmatodendrosis and gliovascular abnormalities in ageing and post-stroke dementia. Brain 139(Pt 1):242–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Early AN, Gorman AA, Van Eldik LJ, Bachstetter AD, and Morganti JM (2020) Effects of advanced age upon astrocyte-specific responses to acute traumatic brain injury in mice. J Neuroinflammation 17(1):115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Yamashita T, Kamiya T, Deguchi K, Inaba T, Zhang H, Shang J, et al. (2009) Dissociation and protection of the neurovascular unit after thrombolysis and reperfusion in ischemic rat brain. J Cereb Blood Flow Metab 29(4):715–725. [DOI] [PubMed] [Google Scholar]
  • 148.Fukuda S, Fini CA, Mabuchi T, Koziol JA, Eggleston LL Jr., and del Zoppo GJ (2004) Focal cerebral ischemia induces active proteases that degrade microvascular matrix. Stroke 35(4):998–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Zlokovic BV (2008) The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 57(2):178–201. [DOI] [PubMed] [Google Scholar]
  • 150.Ivens S, Kaufer D, Flores LP, Bechmann I, Zumsteg D, Tomkins O, et al. (2007) TGF-beta receptor-mediated albumin uptake into astrocytes is involved in neocortical epileptogenesis. Brain 130(Pt 2):535–547. [DOI] [PubMed] [Google Scholar]
  • 151.Piao CS, Holloway AL, Hong-Routson S, and Wainwright MS (2019) Depression following traumatic brain injury in mice is associated with down-regulation of hippocampal astrocyte glutamate transporters by thrombin. J Cereb Blood Flow Metab 39(1):58–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Schachtrup C, Ryu JK, Helmrick MJ, Vagena E, Galanakis DK, Degen JL, et al. (2010) Fibrinogen triggers astrocyte scar formation by promoting the availability of active TGF-beta after vascular damage. J Neurosci 30(17):5843–5854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Sofroniew MV (2020) Astrocyte Reactivity: Subtypes, States, and Functions in CNS Innate Immunity. Trends Immunol 41(9):758–770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Dong Y, and Benveniste EN (2001) Immune function of astrocytes. Glia 36(2):180–190. [DOI] [PubMed] [Google Scholar]
  • 155.Kim RY, Hoffman AS, Itoh N, Ao Y, Spence R, Sofroniew MV, et al. (2014) Astrocyte CCL2 sustains immune cell infiltration in chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 274(1–2):53–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Moreno M, Bannerman P, Ma J, Guo F, Miers L, Soulika AM, et al. (2014) Conditional ablation of astroglial CCL2 suppresses CNS accumulation of M1 macrophages and preserves axons in mice with MOG peptide EAE. J Neurosci 34(24):8175–8185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Cekanaviciute E, Dietrich HK, Axtell RC, Williams AM, Egusquiza R, Wai KM, et al. (2014) Astrocytic TGF-beta signaling limits inflammation and reduces neuronal damage during central nervous system Toxoplasma infection. J Immunol 193(1):139–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Anderson MA, Burda JE, Ren Y, Ao Y, O’Shea TM, Kawaguchi R, et al. (2016) Astrocyte scar formation aids central nervous system axon regeneration. Nature 532(7598):195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, and Sofroniew MV (2004) Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci 24(9):2143–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Herrmann JE, Imura T, Song B, Qi J, Ao Y, Nguyen TK, et al. (2008) STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J Neurosci 28(28):7231–7243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Clarke LE, Liddelow SA, Chakraborty C, Munch AE, Heiman M, and Barres BA (2018) Normal aging induces A1-like astrocyte reactivity. Proc Natl Acad Sci U S A 115(8):E1896–E1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, et al. (2012) Genomic analysis of reactive astrogliosis. J Neurosci 32(18):6391–6410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Brown WR, and Thore CR (2011) Review: cerebral microvascular pathology in ageing and neurodegeneration. Neuropathol Appl Neurobiol 37(1):56–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Perez JM, Evans AC, and Alzheimer’s Disease Neuroimaging I (2016) Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat Commun 7:11934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Korte N, Nortley R, and Attwell D (2020) Cerebral blood flow decrease as an early pathological mechanism in Alzheimer’s disease. Acta Neuropathol 140(6):793–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Salinet AS, Robinson TG, and Panerai RB (2015) Effects of cerebral ischemia on human neurovascular coupling, CO2 reactivity, and dynamic cerebral autoregulation. J Appl Physiol (1985) 118(2):170–177. [DOI] [PubMed] [Google Scholar]
  • 167.Vermeer SE, Hollander M, van Dijk EJ, Hofman A, Koudstaal PJ, Breteler MM, et al. (2003) Silent brain infarcts and white matter lesions increase stroke risk in the general population: the Rotterdam Scan Study. Stroke 34(5):1126–1129. [DOI] [PubMed] [Google Scholar]

RESOURCES