
Research developments in the neurovascular unit and the blood‑brain barrier (Review)
- Authors:
- Published online on: March 18, 2025 https://doi.org/10.3892/br.2025.1966
- Article Number: 88
-
Copyright: © Gong et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
The brain is the most complex organ in the body, and it has a highly specialized structure and function. It accounts for ~2% of body weight, but consumes 20% of the metabolic reserves of the body (1). The metabolic needs of the brain are met through blood transport, and a group of cells from blood vessels and nerves known as the neurovascular unit (NVU) complete the blood supply of the brain (2). The NVU is a complex component that is composed of neurons, astrocytes, oligodendrocytes, microglia, pericytes, and brain microvascular endothelial cells (BMECs) (3-5) (Fig. 1). The blood-brain barrier (BBB) is an important NVU component (6). It is impossible to observe the functional activity of the BBB without the NVU.
The BBB controls the movement of nutrients and metabolites into and out of the brain parenchyma, regulates the brain microenvironment by providing transportation of selective substances, controls molecules and cells, and protects the normal structure and function of the brain (7). As early as 1885, Paul Ehrlich marked all tissues except the brain and spinal cord by injecting aniline stains into the blood vessels (8). Subsequently, Edwin Goldmann successfully stained the brain cells by injecting the dye into the spinal fluid, but other tissues and organs remained intact. This researcher then discovered a special barrier between the central nervous system (CNS) and circulating blood (9). Max Lewandowsky further discovered that capillary obstruction prevented some molecules from entering the brain and introduced the term ‘Bluthirnschranke’ (9). Until the late 19th century, Dr. Lewandowski from Berlin officially named it the BBB (8). It has now been established that the BBB is composed of BMECs, pericytes, astrocytes, and a basement membrane as the barrier matrix (10). The BBB prevents toxic substances, microorganisms, and pathogens in the blood from directly contacting the brain tissue. This protects the brain parenchyma from blood-borne pathogens and prevents drugs and other exogenous compounds from entering the CNS (11,12). Therefore, the BBB function is closely related to stroke (13), traumatic brain injury (TBI) (14), surgical brain injury (SBI) (15), intracerebral hemorrhage (ICH) (16), Alzheimer's disease (17), epilepsy (18), Huntington's disease (17), as well as other neurological diseases.
The present review focuses on the mechanism by which the NVU constructs the BBB. New possibilities for protecting the integrity of the BBB are also provided.
2. Neurons and the BBB
Physiological structure and neuronal functions
The primary function of the nervous system (NS) is to perceive and analyze changes in the internal and external environment of the body to control it (19). Neurons are the basic structures and functional units of the NS that perform the primary functional activities of the NS (20). Neurons are divided into cell bodies and protrusions, and the protrusions contain dendrites and axons (21). Axons comprise >95% of the total volume of neurons, and they carry electrical impulses and project them to other neurons (22). The perivascular neuron axons include the presynaptic portion that is inserted into the basement membrane through the astrocytic endfeet and forms a synaptic-like transmission with vascular smooth muscle cells to regulate high levels of neuronal activity and brain metabolic requirements (23,24). Higher brain functions depend on the electrophysiological signals that are released by neurons. Hence, the BBB is needed to maintain a precise and balanced ionic environment in the brain (23). Neurons form before the BBB during brain development. Neuronal cells combine with other nerve cells to construct the BBB. The BBB, in turn, protects neurons from harmful toxins, pathogens, and immune cells by controlling substance exchanges. Therefore, it is suggested that BBB formation and neuronal development are interrelated and complement each other. Specialized cells that sense the environment have emerged in the simplest multicellular organisms with epithelial tissue (25). These cells may have evolved into the first sensory neurons. As sensory neurons specialize in function, they require the support and protection of other cells. These supporting cells may then evolve into glial cells that form a tight barrier in the brain. This process would promote the separation of neuroactive substances in the central and peripheral areas, helping to establish a consistent microenvironment in the brain (25,26).
Pathophysiological mechanisms of neuronal regulation of the BBB
Neurons are also known as pacemakers of NVUs (27). Neurons convert small changes detected in the surrounding environment into electrical signals and chemical information that are transmitted to neighboring interneurons or astrocytes, thereby affecting vascular tone and regulating BBB permeability (28,29). The necessary regulatory mechanisms are activated to transmit these signals. Neuronal excitability is controlled by calcium and potassium homeostasis, while specific channels and transporters on the BBB provide a favorable environment for synapses and neural activity (30,31). For example, Na+-K+-ATPase plays an important role in cell excitation by regulating the transfer of Na+ and K+ in and out of the brain (32). When ATP production in the brain is insufficient, the Na+-K+-ATPase pump fails, intracellular sodium accumulates, the ion transport gradient collapses, intracellular fluid accumulates, and cells eventually rupture. This causes inflammation and damage to adjacent cells, thus destroying the BBB (33). Voltage-gated Ca2+ channels and K+ channels also regulate ion transport across the BBB (34,35). The normal functioning of the Na+/HCO3--cotransporter and cation-chloride ion cotransporter in neurons also maintains the BBB integrity and intracranial ion homeostasis (36). These transporters or gated channels are expressed in neurons that can regulate and maintain the normal polarity of nerve cells and regulate cell volume and membrane permeability. This contributes to the selective permeability of the BBB to different substances. The permeability of the BBB is reduced for essential water-soluble nutrients and metabolites required by nerve tissue, and these are allowed to pass through to enter the nutrient nerves of the brain. The BBB acts as a specific filtering system for other toxic substances to prevent them from entering the brain (37).
A result of BBB breakdown is cerebral edema, and this can occur in all types of brain cells and brain parenchyma, such as neurons, astrocytes, and endothelial cells (ECs) (38). Following brain injury, neuronal water disintegrates cell bodies and processes, the neuronal nuclei become sparse, the water-soluble cytoplasm disintegrates, and brain edema becomes significantly aggravated (39). Mature nerve cells cannot proliferate. Therefore, the specific function and viability of nerve cells are impaired, and this can lead to BBB damage and further affect brain function (40). This provides strong support for the role of neurons in the BBB. Although the specific pathways of these regulatory mechanisms require further exploration, some signaling pathways have been shown to affect the BBB by disrupting neurons. It was determined from our previous studies that intraperitoneal injection of closantel allows bumetanide to pass through the BBB and inhibits the phosphorylation of the STE20/SPS1-related proline/alanine-rich protein kinase (SPAK)/Na+-K+-Cl- cotransporter 1 (NKCC1) signaling pathway in neurons. This reduces nerve cell apoptosis and has a protective BBB effect (41,42). The SPAK/NKCC1 signaling pathway is activated by phosphorylation after brain injury. Ion channels in the cell membrane open, and Na+, K+, Cl−, and water enter the neuronal cells and disrupt the cellular structure. A large amount of neuronal apoptosis, BBB damage, albumin infiltration into the brain parenchyma, and brain edema of the injured side were found to be significantly aggravated in rats (41,42).
Endoplasmic reticulum stress (ERS) was also found to play an important role in neuronal death. Ca2+ homeostasis of the neuronal ER is disrupted after brain cell injury. This causes ERS that participates in the pathological process of BBB destruction (43). The protein kinase R-like endoplasmic reticulum kinase (PERK) pathway is part of the unfolded protein response (UPR) during ERS. It is activated by phosphorylation after early ICH, and it further phosphorylates downstream eukaryotic translation initiation factor 2α (eIF2α) and upregulates activating transcription factor 4 (ATF4) to play a pro-apoptotic role. Acyl-CoA synthetase long chain family member 4 (ACSL4) reduces Fe3+ to Fe2+ through transferrin and transfers it into cells. The reactive oxygen species (ROS) reaction with Fe2+ leads to ferroptosis, injury of nerve cells, and destruction of the barrier function, causing secondary cerebral edema (44). Deferoxamine can reduce ERS and protect nerve cells by interfering with Fe3+ and inhibiting the PERK signaling pathway (45). However, transmembrane protein 2 (TMEM2) is involved in ERS through non-UPR pathways. TMEM2 decomposes the extracellular high-molecular-weight hyaluronic acid (HMW-HA) into low-molecular-weight (LMW)-HA. The LMW-HA enters cells through the membrane channel, via CD44, inhibiting intracellular p38/ERK signal transduction, and alleviating ERS. Moreover, when TMEM2 is silenced, caspase-3 is highly expressed, and this promotes neuronal apoptosis and cerebral edema (46). In addition, it was determined in our previous study that leucine-rich repeat kinase 2 (LRRK2) inhibition can effectively interfere with Ras-related protein Rab-10 (RAB10) phosphorylation, downregulate lysosomal hydrolase secretion caused by lysosomal overload stress after SBI, and play a protective role in BBB and nerve function (47). Collectively, our previous studies confirmed that the normal structure and function of neurons contribute to BBB integrity maintenance (Fig. 2).
Summary
Neurons are considered to be components of the NVU. Neurons can send signals to regulate the neural network when their energy demands change, and this has direct effects on the BBB. Neurons can rapidly transmit chemical information through synapses, and this may be one of the reasons why neurons respond quickly after BBB damage. In addition, ion concentration differences inside and outside the cell will also initiate the release of membrane potential from the neurons, thereby adjusting the brain microenvironment according to the current demand. It is considered that the mechanism by which neurons participate in BBB regulation is related to their signaling function in the brain, and some new therapeutic targets may emerge by studying this cellular communication.
At present, it is generally considered by most, that it is difficult to regenerate neurons in the adult CNS, and the function of the CNS can be restored after damage because of the compensation of other neurons around it. However, whether the BBB can be damaged by regulating the metabolism of peripheral neurons to play a role in repair is still unknown. The position of neurons is crucial. However, the mechanisms that could explain the direct molecular effects on the integrity of the BBB remain to be determined.
3. Microglia in the BBB
Physiological functions of microglia
Microglia have not yet been determined as direct contributors to BBB maintenance. However, structurally, most para-vascular microglia are located in the perivascular space and are closely related to vascular function (48). Vascular-associated microglia initially maintain the integrity of the BBB by expressing the tight junction (TJ) protein, claudin-5, and make physical contact with ECs (49). Microglia also function through direct contact with neuronal synapses. Microglia monitor their surroundings and continuously extend and retract their cellular protuberance, making direct contact with neuronal synapses at an hourly rate. This continuous and rapid monitoring is a unique function of microglia in the brain (50). In addition, microglia can secrete a variety of cytokines and growth factors, such as brain-derived neurotrophic factor (BDNF), IL-10, and transforming growth factor-β (TGF-β), to help repair local nerve tissue (51,52). These functions of microglia play important roles in restoring brain balance (53). Normally, the CNS is an immunologically privileged region, and the BBB separates the CNS from the inflammatory state to maintain brain homeostasis (54). However, microglia release high levels of inflammatory factors as they are CNS immune cells. Following brain injury, they are the first to be activated among all immune cells to destroy the BBB and participate in the pathological process of neuroinflammation (55,56). The inflammatory environment in the brain is an important factor in BBB destruction. Immune cells cross the BBB and infiltrate the brain parenchyma through paracellular and transcellular pathways, and this leads to extensive neuroinflammation (57). It is known that neuroinflammation is an important driver of numerous diseases and is closely related to the BBB. Proinflammatory cytokines, such as interleukin (IL)-1β, IL-6, and IL-18, tumor necrosis factor (TNF), chemokine ligand (CCL)1 and CCL5, small molecule messengers (prostaglandins, NO, and reactive oxygen species) are produced in large quantities during neuroinflammation. Microglia are one of the main cells involved in this process. As the key cell of brain inflammation, when its immune function is activated, inflammatory factors are released in large quantities. This induces aquaporin 4 (AQP4) upregulation. TJ expression disruption in the ECs affects their functions, leading to vascular disintegration and severe barrier dysfunction (58,59). The damaged barrier displays more leakage, and some of the serum proteins enter the brain to recruit microglia into the blood vessels to phagocytize ECs and destroy the BBB (60). Moreover, serum proteins that leak into brain tissue can bind to local cells and induce secondary brain injury.
Pathophysiological mechanisms of microglia regulation of the BBB
The aforementioned inflammatory effects of microglia are closely related to their morphology. Microglia are activated and polarized into different phenotypes that include M1 (pro-inflammatory effect) and M2 (anti-inflammatory effect) (61). Cerebral edema is one of the most common BBB injury manifestations and can be caused by neuronal cytotoxic edema. Neuronal axons are damaged, and myelin degradation produces a large amount of cellular debris (62). M1 microglia are capable of engulfing these cellular debris, apoptotic neurons, and harmful substances such as inflammatory cytokines and chemokines that are produced during injury (63,64). However, with an increase in M1 microglia, the phagocytic function gradually decreases, inflammatory cells are recruited, and neurotoxic substances accumulate in the brain. This leads to neuronal death and further destruction of the BBB (65,66). NF-κB regulates the expression of most microglia characteristic genes of the M1 phenotype. Peroxisome proliferator activated receptor (PPAR)-γ in activated microglia can inhibit NF-κB signaling to downregulate the microglia ‘M1’ and reduce the expression levels of inducible nitric oxide synthase (iNOS), TNF, IL-1, IL-6, and IL-23 (67-69). In addition, NF-κB has been revealed to promote IL-4 secretion and microglia M2 polarization, thereby improving the tube formation function and cell connection ability of BMECs (70,71). Research has shown that small nucleolar RNA host gene 8 activates microglia to release inflammatory factors to damage BMECs by targeting the sirtuin1 (SIRT1)-mediated NF-κB pathway (72). High mobility group protein B1 interacts with Toll-like receptor 4 to activate the NF-κB/NLRP3 signaling pathway through myeloid differential protein-88 and induce the transcription of proinflammatory cytokines (66). Inhibition of this signaling pathway induces the upregulation of M2 markers, CD206 and arginase 1, and the downregulation of M1 marker proteins, CD86 and iNOS, resulting in the conversion of microglia (73,74). The Notch1/Hes1/Stat3 signaling pathway plays an important role in microglia activation, reprogramming M1 to M2 by inhibiting NF-κB p65 translocation (75). Microglia-mediated inflammation is an important cause of BBB disruption. As the most predominant proinflammatory cell in the NS, microglia also disrupt the BBB by engulfing astrocyte endfeet during persistent inflammation (49).
Recent studies have found that the expression of triggering receptor expressed on myeloid cells 2 (TREM2) in microglia is closely related to BBB destruction after brain injury (76,77). These findings suggest that TREM2 is closely related to the BBB, which is consistent with the results of our study (78). Following brain injury, TREM2 was revealed to be highly expressed in the microglia of the injured side of the brain and negatively correlated with the diffusion of inflammatory factors. TREM2 downregulation resulted in upregulation of NF-κB-mediated inflammatory signals such as IL-6, IL-1β, and TNF-α. With a large amount of nerve cell apoptosis, the BBB was disrupted, albumin exudated into the brain parenchyma surrounding the lesion, and the brain water content on the affected side increased. These results indicated that TREM2 can protect nerve cells and maintain CNS homeostasis (78).
Summary
It was determined that under pathological conditions, microglia morphologies rapidly change to larger cell bodies and shorter processes. This change may be due to a shift in the role of microglia from central monitors to phagocytes. This may have bidirectional effects after BBB disruption. Microglia help remove cellular debris, but they also release inflammatory factors and aggravate neuroinflammation. The benefits and harms of this bidirectional effect of microglia on the BBB cannot be assessed. As important nerve cells, microglia cannot be removed to conduct a holistic study. Thus, the mechanism by which microglia restore the function of the BBB has yet to be determined.
4. Astrocytes in the BBB
Anatomical location and physiological function of astrocytes
As the key point of brain metabolism regulation, astrocytic endfeet are attached to blood vessels and establish a direct interface with the vascular chamber of the NVU. They are closely attached to the basal membrane of blood vessels and form a boundary along the perivascular space, an important part of the BBB (79). Astrocytes are widely distributed in the CNS, and they fill the space outside neurons and blood vessels and secrete neurotrophic factors that support and nourish neurons (80). Astrocytes have isolation and barrier functions. They can prevent neighboring neurons from interfering with each other and have a certain selective effect on the transport of substances, and they participate in BBB formation (81,82). As satellite cells of the NS, astrocytes divide the cortex and gray matter into functional areas. In addition, a single glial cell regulates the internal and external output, guiding nerve axon regeneration (79,83). A single astrocyte forms higher-order glial vascular units along capillaries that match local neural activity and blood flow, coordinate neuronal firing thresholds, and provide metabolic support for neurons and their synapses (84). Astrocytic endfeet extend to the periphery of brain microvessels, control the barrier function of BMECs through derived factors and physical contact, and regulate the BBB bidirectionally (85).
Astrocytes can divide and proliferate, unlike neurons. A damaged or degenerated brain primarily relies on astrocyte proliferation to fill the tissue defect. It has been confirmed that astrocytes can be induced to differentiate into neurons in vitro, and this provides hope for the treatment of a variety of neurodegenerative diseases (86). It has been observed that altered astrocyte communication can induce neuronal apoptosis and phagocytosis of broken neurons, and this has direct consequences for neuropathology (87,88). However, astrocytes detect glutamatergic and gabaergic neuronal signals and translate them into commands to expand/contract blood vessels (89). They promote the transmission of ions, amino acids, neurotransmitters, and other substances in the brain by absorbing and regulating nerve molecules, and they also establish water homeostasis in the brain (90,91). Thus, they regulate the local microenvironment and affect the extent of BBB damage and subsequent repair. Astrocytes can stabilize K+ and H+ concentrations in the extracellular fluid of the CNS during normal brain activity (92). Water is drained from the brain and out of the body, while neurotransmitters and ions are recycled. When proliferating astrocytes produce scarring, the absorption capacity for K+ and H+ is correspondingly weakened. This results in high K+ in the local extracellular fluid. These boundary and scar functions play important roles in maintaining the physiological homeostasis of the CNS, supporting nerve function and signal transduction (93,94). Following brain injury, the morphology of astrocytes changes from static to reactive. This morphological change affects the BBB and induces brain pathology (95,96). In summary, astrocytes support neuronal cells by regulating neurofactors and preventing hemodynamic and metabolic disorders. They create conditions for the survival and recovery of neurons, protect the permeability integrity of the BBB, and alleviate secondary brain injury.
There is increasing evidence that astrocytes play a key role in BBB breakdown after brain injury. There are abundant organic anion transporters (OAPs) in the contact interface between the astrocytic endfeet and a blood vessel. When this contact is broken, the density of OAPs, such as Kir4.1 and AQP4, that are carried by the astrocytic endfeet decreases. The ion and water imbalances cannot maintain the BBB function, cell edema increases, the cytoskeleton is destroyed, and the brain is damaged further (97,98). Astrocytes secrete vascular permeability factors that enhance the BBB permeability, including vascular endothelial growth factor (VEGF) (99), NO (100), glutamic acid 69, matrix metalloproteinases (MMPs) (101), and endothelin (102).
Pathophysiological mechanisms of astrocyte regulation of the BBB
Astrocytes can generate retinoic acid (103), sonic Hedgehog (Shh) (104), angiopoietin-1 (Ang-1) (105), glia-derived neurotrophic factor (106), insulin-like growth factor (107), and apolipoprotein E (108) to help restore the BBB. Numerous factors released by astrocytes have matching receptors on the brain endothelium that can transmit neuronal activity signals to the ECs and improve the efficiency of nutrition and metabolism of the brain endothelium (109). Genes involved in BBB regulation have also been found in perivascular astrocytes, including integrin subunit α7 (Itga7) (110). Itga7 promotes EC junction integrity by adhering to laminin. Knockdown of Itga7 downregulates fibronectin expression, a major component of the brain barrier. This results in a significant reduction in astrocyte coverage around brain microvessels (111,112). Therefore, how does Itga7 work? Studies have shown that Itga7 activates STAT3 via TNF and increases the expression of serpin family A member 3 encoding the α1-anticoagulant trypsin protein. This prompts astrocytes to enter an inflammatory state (110,113). However, triglyceride-rich lipoprotein (TGRL) liposomes activate the ER protein, CHOP, and NF-κB pathways through the JNK/cJUN/ATF3 pathway to further aggravate inflammatory factor secretions (114). Growth differentiation factor 15 (GDF15) is an important factor in astrocyte remodeling, and it is related to the enhancement of tight BBB connections. TGRL lipolysis products activate GDF15 in astrocytes. Increased GDF15 expression promotes astrocyte remodeling and enhances barrier properties between ECs via claudin-5(115). Astrocyte-derived TGF-β may maintain BBB function by regulating Hedgehog signaling in BMECs (116). TGF-β signal overactivation is a necessary and sufficient condition to cause neurological dysfunction and BBB-related pathology (117). With further research, cellular metabolism was found to be involved in these coordination processes. For example, high blood glucose inhibits astrocyte activation and destroys endfeet. This increases neutrophil infiltration in the brain, leading to BBB leakage (118). Hypertonic saline alleviates BBB permeability by inhibiting the downregulation of VEGF expression by NF-кB p65(119). Intracellular high iron levels regulate BMECs by increasing hepcidin and reducing the uptake of iron from the circulation by the BBB (120).
Several receptors have been found on brain ECs and astrocytes to cause an increase in intracellular Ca2+ upon activation. Upregulation of Ca2+ regulates the activities of neurons, astrocytes, and ECs (121,122). When Ca2+ ions are activated, they send signals from the CNS nerve cells to neighboring cells through the astrocytes. This releases a signal to dilate the vascular space, and a TJ is opened (123,124). Perivascular astrocyte processes regulate the TJs around brain ECs. This pattern of signal transduction between neuroglia and ECs is of great significance for the physiological and pathological regulation of the BBB by brain ECs. Several astrocyte-derived factors have been found to disrupt vascular permeability by decreasing the endothelial TJ protein expression or inducing EC apoptosis. VEGFA expression is a key driver of BBB permeability, and astrocytes drive BBB opening by VEGFA (125). Upregulation of Apelin-13 promotes the phosphorylation of ERK, Akt, and AQP4 in astrocytes (126). Following the upregulation of Apelin-13/APOE4 and NF-κB/HIF-1α signals, MMP9 and VEGF secretions also increase. The reduction of astrocytes endfeet that cover blood vessels and TJ damage are evidence of BBB damage (127,128). Lee et al (129) further found that src-suppressed C-kinase substrate reduced VEGF by decreasing Apelin-1 while stimulating the upregulation of Ang-1. This regulates cerebral angiogenesis and TJs between cells, thereby affecting BBB differentiation (129). Proteins such as astrocytic N-myc downstream regulatory gene 2 and thrombospondin-2 also act as regulators of BBB permeability. They play roles in BBB permeability and immune cell infiltration regulation by affecting MMP expression (130,131).
Astrocytes are often one of the major cell types that initiate the inflammatory cascade. Astrocyte swelling is considered to be an early sign of cytotoxic edema. The inflammatory environment caused by brain injury mediates astrocyte signaling, and this is an important cause of cellular swelling (132). The degradation of the extracellular matrix (ECM) and TJ proteins activates astrocytes, stimulates glial cells to release inflammatory factors that aggravate neuroinflammation, disrupts the TJs of vascular ECs, and accelerates BBB permeability (42,133). Collagen-IV is a major BBB basement membrane component, and its degradation can lead to basement membrane dysfunction. Our previous study showed that MMP-9 was secreted by astrocytes after a TBI (133). This degraded endothelial TJ proteins and the ECM, including occludin and collagen-IV. It was also determined that in addition to directly destroying basement membrane and TJ proteins, MMP-9 also affects TJs by inhibiting the Hedgehog pathway. MMP-9 is upregulated after brain injury, and it partially blocks the Hedgehog pathway and reduces Scube2 and Shh expression levels in astrocytes. This causes further damage to the BBB. Inhibition of this signaling pathway has a protective effect on the BBB mechanism (133,134).
Summary
It is considered that the contribution of astrocytes to the BBB is primarily due to their supporting, nutritional, and other auxiliary effects on nerve cells. Astrocytes can secrete a variety of neurotrophic factors to promote the repair or activation of ECs, BMECs, and pericytes so that they provide better barrier characteristics. Furthermore, astrocytes adhere to the wall of microvessels, encase parts of the microvessels, and strengthen the basement membrane. All of these functions play important roles in maintaining BBB homeostasis. However, the role of astrocytes is much more than this. Astrocytes are also able to release glutamate and thus participate in electrical signaling in the NS. They have an important physiological role in the NS, and this also provides new insights for the treatment of complex neurological diseases.
Astrocytes have attracted notable attention due to their pivotal role in the NVU and have been studied as aforementioned. BBB homeostasis imbalance has been implicated in numerous CNS diseases. The role and mechanism of astrocytes in maintaining BBB homeostasis, especially in the transition from normal BBB homeostasis to imbalances of BBB homeostasis in disease states, require further investigations.
5. BMECs in the BBB
Physiological structure and BMEC functions
The BBB is a selective barrier formed by ECs that line the brain microvessels. As a key NVU regulator, BMECs are key cells for BBB integrity (135). BMECs are a single layer of flat ECs that line the inner surface of cerebral blood vessels. BMECs have a more obese appearance than ECs from other non-neural tissues (136). BMECs have some unique features, such as the presence of numerous intercellular TJs that generate high transendothelial impedance and delay paracellular flux. They lack the fenestra structure of ECs and have low levels of pinocytosis of the liquid phase material (137). In addition, BMECs possess asymmetrically localized enzymes and vector-mediated transport systems.
These characteristics make the connection between two cells tighter, forming a liquid impenetrable barrier that prevents molecules and ions from passing through the intercellular space (37,138). The basement membrane of brain microvessels is intact, and a few phagocytic vesicles are distributed in the vascular lumen. In addition, the number of mitochondria is high (139). BMEC mitochondria account for approximately one-tenth of the cytoplasmic volume, a proportion much higher than that of ECs in other non-BBB tissues (140,141). The high mitochondria proportion helps to improve the efficiency of metabolism and transport, enhance the signal transmitted by sensing neurons, and promote the propagation of vasodilator signals to the tiny arteries in the brain to regulate the vasodilation function (142). BMECs can also transport oxygen and glucose from the blood to the brain to increase its energy supply while emitting carbon dioxide and other waste products in the opposite direction (143,144). This is essential to maintain the physiological function of the NVU and regulate the pH of normal brain metabolism, and is one of the reasons for the sensitivity of the BBB to oxidative stress (145). BMECs can form a tighter capillary endothelium than peripheral ECs, and this aspect renders the BBB essentially impermeable to macromolecules (135). The TJs fill the gaps between BMECs and limit paracellular permeability by expressing TJ proteins such as occludin, claudins, and zonula occludens. These suggest that the TJs of BMECs are indispensable for a normal structure and function of the BBB. The TJs connect adjacent cells and seal intercellular spaces, and this prevents top and bottom membrane proteins from spreading laterally (146,147). The purpose is to block the paracellular fusion pathway of ECs to form a physical barrier. Hydrophobic molecules are allowed to diffuse internally, whereas hydrophilic molecules are hindered by barriers, thus maintaining CNS homeostasis (148). Therefore, drugs that target nerve regions or nerve cells must have the ability to cross the BBB to achieve the desired therapeutic effect (149). BMECs contain transporters, such as ATP synthase-binding cassette (ABC), ABC subfamily member 1, and solute carrier organic anion transporter family member 1C1, as well as receptors, such as endothelial protein C receptor, insulin receptors, transferrin receptors (TfRs), and LDL receptor related protein 1, that can help CNS drugs open the BBB (150). Multiple receptors on BMECs enable cells to respond to inflammatory mediators (151). One of the hallmarks of vascular inflammation is the increased expression of adhesion molecules on the BMEC membrane. Vascular inflammation can affect the infiltration of peripheral immune cells, activate microglia, change the shape of blood vessels, affect angiogenesis, and ultimately destroy blood vessels (152,153).
Pathophysiological mechanisms of BMEC regulation of the BBB
The barrier function of BMECs is maintained through close association with other cell types within the NVU. Direct contact between ECs and astrocytes is necessary for BBB formation and functioning (154). When co-cultured with astrocytes, endothelial soluble factors showed higher transendothelial resistance (155). Changes in ECs and astrocytes were also found in cerebral cortex biopsy specimens from patients with brain injuries (156). This may have been related to EC morphological changes. The injured EC surfaces showed longitudinal folds and invagination, the activity was enhanced, and granulation and some multivesicular bodies were observed in the cytoplasm. The space between ECs and astrocytes was significantly enlarged (157). The interaction between astrocytes and ECs involves intercellular and intracellular communication, which is key to BBB induction and maintenance (84,158). Research has found that autophagy related 7 (Atg7) regulates the function of astrocyte adhesion to the BMECs to maintain BBB homeostasis through endothelial fibronectin (159). Atg7 affects the phosphorylation of the cyclic adenosine monophosphate-responsive element-binding protein by regulating protein kinase A activity, thereby triggering endothelial fibronectin expression. Loss of endothelial Atg7 downregulates fibronectin expression, a component of the BBB, and reduces the extent of astrocyte coverage of cerebral microvessels (159). Other signaling mechanisms between ECs and the BBB have also been gradually discovered. Among them, the VEGF/VEGF receptor (VEGFR) pathway, Notch, and Wnt pathway are considered to be the key signaling pathways that drive the phenotype of BMECs during BBB formation (160). The VEGF/VEGFR signaling pathway is related to the proliferation, invasion, migration, and survival of ECs and is involved in the activation of most mechanisms of angiogenesis (161). The endothelial nitric oxide synthase (eNOS)/NO pathway regulates vascular remodeling, and inhibition of this signaling pathway destroys VEGF. This leads to impaired angiogenesis (162). The eNOS/NO pathway in ECs is related to BDNF. After the impairment of this pathway, BDNF is downregulated, and this affects neurotransmitter release, thereby impairing neuronal regeneration and leading to BBB leakage (163). It is worth noting that the eNOS/NO pathway in BMECs is closely aligned with mitochondria-associated oxidative stress, and as a downstream effector of VEGF, the reduction of NO impairs the VEGF/VEGFR pathway (164). Wnt signaling is also essential for CNS angiogenesis and BBB formation (165). In addition, Notch signaling is involved in EC proliferation and survival processes (166). SIRT1 promotes angiogenesis by promoting VEGFR-2 expression through Notch signaling (167). Taken together, BBB integrity depends to a large extent on the interaction of BEMCs with other cells in the NVU.
VEGF is a growth factor that induces angiogenesis and increases vascular permeability, and it has a key role in BBB integrity loss (168). The blocking of VEGF was demonstrated to alleviate the degradation of the TJ protein occludin (169). Collagen-IV is a major BBB basement membrane component, and its degradation can lead to dysfunction of the basement membrane; this may be one of the causes of BBB disruption after a brain injury (170). Our results, in a previous study showed that in the brain tissues surrounding the TBI in rats, VEGF expression increased, and occludin and collagen-IV expression levels decreased. The basement membrane and TJs of the BBB were disrupted. The TUNEL and Fluoro Jade C-positive cell levels increased, nerve cells were apoptotic, and the BBB was impaired. VEGF inhibition protected the BBB and reduced TBI-induced brain edema. This effect may be achieved through MMP9 regulation of occludin and collagen-IV (133).
Summary
As one of the major cell types constituting the BBB, the contribution of BMECs to the BBB originates from their structural location. BMECs form the BBB through TJs and gap junctions that prevent the passage of macromolecular substances. Of course, in addition to forming barriers, BMECs also participate in a variety of neural metabolism activities and release neuroactive substances that affect neural function. These mechanisms all serve to further regulate the brain microenvironment and protect the BBB.
6. Pericytes in the BBB
Physiological structure and functions of pericytes
Pericytes were discovered as early as 1873 by French scientist Charles Benjamin Rouget, and they were originally called ‘Rouget's’ cells. They were later officially named ‘pericytes’ by Karl Wilhelm Zimmermann (171). Pericytes are an important component of the NVU, and they attach to the lateral wall of the lumen of ECs in capillaries, embed into ECs, and cover approximately one-third of the capillary wall. Together with vascular ECs, they constitute the cellular component of the blood vessel wall and play an important role in the BBB (172). Within the NVU, pericytes are located in a unique location between neurons, astrocytes, and ECs, and their coverage is inversely proportional to the permeability of the blood vessels (173). The cytoplasm of pericyte stellate processes is wrapped outside the vascular basement membrane and is primarily distributed at the TJs of ECs. ECs are surrounded by pericytes and communicate directly with the microvascular endothelium through physical contact and paracrine signals. The structural substances constituting the basement membrane and the ECM, including integrins, N-cadherin, fibronectin, and connexin, regulate capillary permeability and participate in the perfusion and infiltration of microcirculation (171). Pericytes have contractility similar to smooth muscle cells and can regulate cerebral vessel formation and maturation (174). The cerebral angiogenesis signaling pathway is an important regulator that maintains the normal structure of the BBB, and its signaling pathway abnormality can lead to an aberrant BBB structure. Studies have shown that pericytes express receptors for vascular mediators, such as catecholamines, β-adrenoceptors, thromboxane A2, vasoactive intestinal peptide, endothelin-1, and vasopressin, that play crucial roles in maintaining vascular wall stability (175,176). Peripheral cells also participate in angiogenesis by expressing TGF-β, VEGF, Ang-1/2, angiotensin I, and other vascular-related factors (177). Type IV collagen, polysaccharides, and laminin synthesized by pericytes are also important for basement membrane formation. TfR1 is one of the known markers of the BBB, and TfR1 is downregulated in ECs lacking pericellular contact (178). It has been shown that capillary permeability increases after pericyte depletion. This phenomenon supports that pericytes have an important role in maintaining endothelial barrier function (179).
Pathophysiological mechanisms of pericyte regulation of the BBB
Pericytes share a basement membrane with ECs as they can regulate microcirculatory blood flow. Direct synapse-like contacts are formed with ECs via N-cadherin and connexin (180). The interaction between pericytes and BMECs is essential for basement membranes and TJs and directly affects BBB permeability. Gap junctions, focal adhesion, and nail hole junctions are three ways in which pericytes interact with ECs (181). Gap junctions transmit ionic currents and small molecules between neighboring cells (182). Focal adhesion kinase was revealed to immobilize capillary pericytes on ECs (183). Pericytes play a key role in BBB homeostasis by communicating with ECs through these specialized roles (184). Daneman et al (185) found that ECs co-cultured with pericytes had reduced intercellular spaces and increased transepithelial electrical resistance. Abundant TJ proteins that included occludin and claudin 5 were present at the cell boundary, suggesting that the pericytes enhanced the TJ function (185). ECs associated with pericytes are more resistant to apoptosis than ECs alone and have a protective effect on ECs. This provides new evidence that pericytes together with ECs support BBB structural integrity. The phagocytic function of ECs is enhanced after pericyte depletion. Vascular permeability genes, such as Angpt2, Plvap, and LAM, were increased, and molecular transporters were overexpressed. Some of the macromolecules are allowed to cross the BBB to affect the brain parenchyma (186). In addition, the receptor, PDGFR, expressed by pericytes can respond rapidly after brain injury, mediate pericyte migration to ECs, release fibrinolytic enzymes, and increase BBB permeability (187). It is worth exploring that in the absence of pericytes, embryonic brain ECs can still express some BBB-specific molecules. This may be induced by neural progenitor cells that then regulate the functional integrity of the BBB during development through pericytes and adult astrocytes (188).
Pericytes are an important part of brain capillaries that have a phagocytic function and can remove cell debris and regulate capillary blood flow and BBB permeability. Pericytes in different vascular beds have different frequencies and can transform into multipotent vascular stem cells that generate microglia in the late stage of brain injury and participate in microglia proliferation to acquire a microglial phenotype with a phagocytic capacity (189,190). Pericytes can also direct astrocytic endfeet into the endothelium (191). The interaction between pericytes, ECs, and astrocytes can regulate angiogenesis and hemodynamics and maintain BBB integrity. This is essential for normal brain function.
Currently, pericyte loss is considered to be a hallmark of BBB dysfunction, and it causes a variety of secondary brain injuries such as brain edema and brain parenchymal lesions (192). Oxidative stress, a high lipid and high glucose internal environment, and toxic substances in the blood can damage pericytes. This then destroys the BBB. Aβ is the most widely studied of the toxic substances that cause BBB damage. Aβ binds to RAGE to increase oxidative stress, promoting Aβ accumulation in the brain. This process causes BMEC cytotoxicity and induces pericyte ferroptosis (193-195).
Summary
The role of pericytes in the BBB has been further revealed with increasing research. Pericytes are located lateral to the BMECs and control the endothelial function through direct contact to maintain BBB stability. In addition, pericytes are involved in physiological processes such as substance transport and signal transmission to ensure normal brain function. Understanding pericyte functions helps us to better understand BBB processes and their importance in protecting the brain from harmful agents.
7. Conclusion
The normal functioning of the brain depends on the interactions between the components of the NVU. The positive role of these cells in CNS homeostasis is encouraging. Neurons, glial cells, pericytes and vascular ECs constitute the BBB, which provides a stable internal environment for the realization of nerve function. Among them, astrocytes are closely related to neuronutrition and repair, and it is speculated that astrocytes may also have some benign effects on nerve cells and brain microvascular diseases, such as the promotion of microvascular and nerve injury regeneration to help BBB function repair. However, further studies are required to confirm this. The focus on the relationship between neuroinflammation and the BBB has uncovered a notable phenomenon. Activated microglia and reactive astrocytes communicate and cooperate with each other in the NVU to achieve immune ‘optimization’. Brain inflammatory signals are amplified not only by microglial activation but also by the unique anatomy of astrocytes in the immune network (196,197). Microglia and astrocyte interactions may become an effective and accurate therapeutic target in the future with further technological advances. These NVU cell-to-cell interactions are critical to regulate cerebrovascular functions. BBB permeability alterations after acute CNS injury are due to a loss or alteration of these cellular structures, and understanding the structure and mechanisms of these cells is important to control injury progression.
Various NVU components and their interactions are essential to maintain BBB stability. The damage of the BBB is the pathological basis of vascular cerebral edema. The destruction of the BBB can lead to brain edema and increased intracranial pressure, which in turn can aggravate brain edema and cause functional and structural damage to brain tissue. If the brain edema progresses from localized to diffuse, irreversible secondary pathological changes are formed, and even brain death is developed. Brian edema is often secondary to neurological diseases such as TBI, intracranial space occupying lesions, cerebrovascular diseases, cerebral parasitosis and some cerebral congenital diseases. The severity of cerebral edema is related to the course and prognosis of intracranial diseases and has important significance. In the field of neurology, if we can better understand the mechanism of the BBB and effectively prevent and treat the secondary brain edema, we can achieve better results in the treatment of intracranial diseases. The therapeutic potential of the BBB in a variety of neurological diseases has great potential. Hence, it is necessary to investigate the effect of the NVU on the BBB as a whole to further reveal possible therapeutic mechanisms.
The recovery of the barrier function is considered to be the key to the treatment of NS diseases. An exploration of the NVU composition and its cell-cell interactions will help to further unlock the mechanism of the BBB and provide new insights for the clinical treatment of NS diseases. Drug therapy should protect the whole NVU from different angles and in various ways. The exploration of multi-target protective agents has become one of the important research and development directions for the treatment of NS diseases in the future.
In conclusion, it is considered that the NVU should be regarded as a functional whole, and the dynamic interaction between all cells affects brain function. However, most of the current BBB studies have only focused on a single nerve cell or target, and few have studied a variety of nerve cells and brain microvascular endothelium as a whole. In the future, focus will be on the cellular structure and function of the NVU, and exploration on how a variety of cells combine to repair the BBB after disruption.
Acknowledgements
Not applicable.
Funding
Funding: The present review was supported by the National Natural Science Foundation of China (grant no. 82205254), the 11th Batch of Science and Technology Development Projects of Suzhou in 2023 (grant no. SKY2023014), the University-local Collaborative Innovation Research Project (grant no. 20239606), the Jiangsu Traditional Chinese Medicine Science and Technology Development Program project (grant no. MS2023101) and the Wumen Medical Project of Suzhou (grant no. SKYXD2022034).
Availability of data and materials
The literature analyzed during the current study are available in the PubMed (https://pubmed.ncbi.nlm.nih.gov/), Embase (https://www.embase.com/), Ovid (https://ovidsp.ovid.com/), CNKI (https://www.cnki.net/) and Cochrane Library (https://www.cochranelibrary.com/library) repositories.
Authors' contributions
BD and YG conceived and designed the manuscript. MW, XH and YH searched the literature and wrote the manuscript. JY created the figures. BD reviewed and revised the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Diaz-Garcia CM, Mongeon R, Lahmann C, Koveal D, Zucker H and Yellen G: Neuronal stimulation triggers neuronal glycolysis and not lactate uptake. Cell Metab. 26:361–374.e4. 2017.PubMed/NCBI View Article : Google Scholar | |
Roy CS and Sherrington CS: On the regulation of the blood-supply of the brain. J Physiol. 11:85–158.117. 1890.PubMed/NCBI View Article : Google Scholar | |
Xue Q, Liu Y, Qi H, Ma Q, Xu L, Chen W, Chen G and Xu X: A novel brain neurovascular unit model with neurons, astrocytes and microvascular endothelial cells of rat. Int J Biol Sci. 9:174–189. 2013.PubMed/NCBI View Article : Google Scholar | |
Harder DR, Zhang C and Gebremedhin D: Astrocytes function in matching blood flow to metabolic activity. News Physiol Sci. 17:27–31. 2002.PubMed/NCBI View Article : Google Scholar | |
Simard M, Arcuino G, Takano T, Liu QS and Nedergaard M: Signaling at the gliovascular interface. J Neurosci. 23:9254–9262. 2003.PubMed/NCBI View Article : Google Scholar | |
Reese TS and Karnovsky MJ: Fine structural localization of a blood-brain barrier to exogenous peroxidase. J Cell Biol. 34:207–217. 1967.PubMed/NCBI View Article : Google Scholar | |
Yamamizu K, Iwasaki M, Takakubo H, Sakamoto T, Ikuno T, Miyoshi M, Kondo T, Nakao Y, Nakagawa M, Inoue H and Yamashita JK: In vitro modeling of blood-brain barrier with human iPSC-derived endothelial cells, pericytes, neurons, and astrocytes via notch signaling. Stem Cell Reports. 8:634–647. 2017.PubMed/NCBI View Article : Google Scholar | |
Ehrlich P: Das sauerstoff-bedürfnis des organismus: Eine farbenanalytische studie. August Hirschwald, 1885. | |
Saunders NR, Dreifuss JJ, Dziegielewska KM, Johansson PA, Habgood MD, Møllgård K and Bauer HC: The rights and wrongs of blood-brain barrier permeability studies: A walk through 100 years of history. Front Neurosci. 8(404)2014.PubMed/NCBI View Article : Google Scholar | |
Patel R, Page S and Al-Ahmad AJ: Isogenic blood-brain barrier models based on patient-derived stem cells display inter-individual differences in cell maturation and functionality. J Neurochem. 142:74–88. 2017.PubMed/NCBI View Article : Google Scholar | |
Salmina AB, Kharitonova EV, Gorina YV, Teplyashina EA, Malinovskaya NA, Khilazheva ED, Mosyagina AI, Morgun AV, Shuvaev AN, Salmin VV, et al: Blood-brain barrier and neurovascular unit in vitro models for studying mitochondria-driven molecular mechanisms of neurodegeneration. Int J Mol Sci. 22(4661)2021.PubMed/NCBI View Article : Google Scholar | |
Salmina AB, Kuvacheva NV, Morgun AV, Komleva YK, Pozhilenkova EA, Lopatina OL, Gorina YV, Taranushenko TE and Petrova LL: Glycolysis-mediated control of blood-brain barrier development and function. Int J Biochem Cell Biol. 64:174–184. 2015.PubMed/NCBI View Article : Google Scholar | |
Lv J, Hu W, Yang Z, Li T, Jiang S, Ma Z, Chen F and Yang Y: Focusing on claudin-5: A promising candidate in the regulation of BBB to treat ischemic stroke. Prog Neurobiol. 161:79–96. 2018.PubMed/NCBI View Article : Google Scholar | |
Zhu J, Lin X, Chen C, Tan H, Gao Y, Li D and Chen G: WNK3 promotes neuronal survival after traumatic brain injury in rats. Neuroscience. 477:76–88. 2021.PubMed/NCBI View Article : Google Scholar | |
Huang W, Li J, Geng X, Li S, Zou Y, Li Y, Jing C and Yu H: The reactive astrocytes after surgical brain injury potentiates the migration, invasion, and angiogenesis of C6 glioma. World Neurosurg. 168:e595–e606. 2022.PubMed/NCBI View Article : Google Scholar | |
Wu D, Lai N, Deng R, Liang T, Pan P, Yuan G, Li X, Li H, Shen H, Wang Z and Chen G: Activated WNK3 induced by intracerebral hemorrhage deteriorates brain injury maybe via WNK3/SPAK/NKCC1 pathway. Exp Neurol. 332(113386)2020.PubMed/NCBI View Article : Google Scholar | |
Drouin-Ouellet J, Sawiak SJ, Cisbani G, Lagacé M, Kuan WL, Saint-Pierre M, Dury RJ, Alata W, St-Amour I, Mason SL, et al: Cerebrovascular and blood-brain barrier impairments in Huntington's disease: Potential implications for its pathophysiology. Ann Neurol. 78:160–177. 2015.PubMed/NCBI View Article : Google Scholar | |
Mohi-Ud-Din R, Mir RH, Mir PA, Banday N, Shah AJ, Sawhney G, Bhat MM, Batiha GE and Pottoo FH: Dysfunction of ABC transporters at the surface of BBB: Potential implications in intractable epilepsy and applications of nanotechnology enabled drug delivery. Curr Drug Metab. 23:735–756. 2022.PubMed/NCBI View Article : Google Scholar | |
Ferraro S, Klugah-Brown B, Tench CR, Bazinet V, Bore MC, Nigri A, Demichelis G, Bruzzone MG, Palermo S, Zhao W, et al: The central autonomic system revisited-convergent evidence for a regulatory role of the insular and midcingulate cortex from neuroimaging meta-analyses. Neurosci Biobehav Rev. 142(104915)2022.PubMed/NCBI View Article : Google Scholar | |
Bittern J, Pogodalla N, Ohm H, Brüser L, Kottmeier R, Schirmeier S and Klämbt C: Neuron-glia interaction in the Drosophila nervous system. Dev Neurobiol. 81:438–452. 2021.PubMed/NCBI View Article : Google Scholar | |
Rolls MM and Jegla TJ: Neuronal polarity: An evolutionary perspective. J Exp Biol. 218:572–580. 2015.PubMed/NCBI View Article : Google Scholar | |
Muzio MR and Cascella M: Marco Cascella declares no relevant financial relationships with ineligible companies. In: Histology, Axon. StatPearls, Treasure Island, FL, 2024. | |
Zhang D, Ruan J, Peng S, Li J, Hu X, Zhang Y, Zhang T, Ge Y, Zhu Z, Xiao X, et al: Synaptic-like transmission between neural axons and arteriolar smooth muscle cells drives cerebral neurovascular coupling. Nat Neurosci. 27:232–248. 2024.PubMed/NCBI View Article : Google Scholar | |
Qin D and Wang J, Le A, Wang TJ, Chen X and Wang J: Traumatic brain injury: Ultrastructural features in neuronal ferroptosis, glial cell activation and polarization, and blood-brain barrier breakdown. Cells. 10(1009)2021.PubMed/NCBI View Article : Google Scholar | |
Schirmeier S and Klämbt C: The Drosophila blood-brain barrier as interface between neurons and hemolymph. Mech Dev. 138:50–55. 2015.PubMed/NCBI View Article : Google Scholar | |
Miller F, Afonso PV, Gessain A and Ceccaldi PE: Blood-brain barrier and retroviral infections. Virulence. 3:222–229. 2012.PubMed/NCBI View Article : Google Scholar | |
Cataldi M: The changing landscape of voltage-gated calcium channels in neurovascular disorders and in neurodegenerative diseases. Curr Neuropharmacol. 11:276–297. 2013.PubMed/NCBI View Article : Google Scholar | |
Lauritzen M, Mathiesen C, Schaefer K and Thomsen KJ: Neuronal inhibition and excitation, and the dichotomic control of brain hemodynamic and oxygen responses. Neuroimage. 62:1040–1050. 2012.PubMed/NCBI View Article : Google Scholar | |
Cheng YT, Luna-Figueroa E, Woo J, Chen HC, Lee ZF, Harmanci AS and Deneen B: Inhibitory input directs astrocyte morphogenesis through glial GABA(B)R. Nature. 617:369–376. 2023.PubMed/NCBI View Article : Google Scholar | |
Ancatén-González C, Segura I, Alvarado-Sánchez R, Chávez AE and Latorre R: Ca2+- and voltage-activated K+ (BK) channels in the nervous system: One gene, a myriad of physiological functions. Int J Mol Sci. 24(3407)2023.PubMed/NCBI View Article : Google Scholar | |
Cohen S and Greenberg ME: Communication between the synapse and the nucleus in neuronal development, plasticity, and disease. Annu Rev Cell Dev Biol. 24:183–209. 2008.PubMed/NCBI View Article : Google Scholar | |
Sun J, Zheng Y, Chen Z and Wang Y: The role of Na+ -K+ -ATPase in the epileptic brain. CNS Neurosci Ther. 28:1294–1302. 2022.PubMed/NCBI View Article : Google Scholar | |
Jiang S, Fan F, Yang L, Chen K, Sun Z, Zhang Y, Cairang N, Wang X and Meng X: Salidroside attenuates high altitude hypobaric hypoxia-induced brain injury in mice via inhibiting NF-κB/NLRP3 pathway. Eur J Pharmacol. 925(175015)2022.PubMed/NCBI View Article : Google Scholar | |
Li B, Li N, Chen L, Ren S, Gao D, Geng H, Fu J, Zhou M and Xing C: Alleviating neuroinflammation through photothermal conjugated polymer nanoparticles by regulating reactive oxygen species and Ca2+ signaling. ACS Appl Mater Interfaces. 14:48416–48425. 2022.PubMed/NCBI View Article : Google Scholar | |
Reddiar SB, de Veer M, Paterson BM, Sepehrizadeh T, Wai DCC, Csoti A, Panyi G, Nicolazzo JA and Norton RS: A biodistribution study of the radiolabeled Kv1.3-blocking peptide DOTA-HsTX1[R14A] demonstrates brain uptake in a mouse model of neuroinflammation. Mol Pharm. 20:255–266. 2023.PubMed/NCBI View Article : Google Scholar | |
Carbajal-Contreras H, Murillo-de-Ozores AR, Magaña-Avila G, Marquez-Salinas A, Bourqui L, Tellez-Sutterlin M, Bahena-Lopez JP, Cortes-Arroyo E, Behn-Eschenburg SG, Lopez-Saavedra A, et al: Arginine vasopressin regulates the renal Na+-Cl- and Na+-K+-Cl- cotransporters through with-no-lysine kinase 4 and inhibitor 1 phosphorylation. Am J Physiol Renal Physiol. 326:F285–F299. 2024.PubMed/NCBI View Article : Google Scholar | |
Engelhardt B and Sorokin L: The blood-brain and the blood-cerebrospinal fluid barriers: Function and dysfunction. Semin Immunopathol. 31:497–511. 2009.PubMed/NCBI View Article : Google Scholar | |
Stokum JA, Gerzanich V and Simard JM: Molecular pathophysiology of cerebral edema. J Cereb Blood Flow Metab. 36:513–538. 2016.PubMed/NCBI View Article : Google Scholar | |
Wiley CA, Bissel SJ, Lesniak A, Dixon CE, Franks J, Beer Stolz D, Sun M, Wang G, Switzer R, Kochanek PM and Murdoch G: Ultrastructure of diaschisis lesions after traumatic brain injury. J Neurotrauma. 33:1866–1882. 2016.PubMed/NCBI View Article : Google Scholar | |
Hosokawa M and Ueno M: Aging of blood-brain barrier and neuronal cells of eye and ear in SAM mice. Neurobiol Aging. 20:117–123. 1999.PubMed/NCBI View Article : Google Scholar | |
Gong Y, Wu M, Gao F, Shi M, Gu H, Gao R, Dang BQ and Chen G: Inhibition of the p-SPAK/p-NKCC1 signaling pathway protects the blood-brain barrier and reduces neuronal apoptosis in a rat model of surgical brain injury. Mol Med Rep. 24(717)2021.PubMed/NCBI View Article : Google Scholar | |
Gong Y, Wu M, Shen J, Tang J, Li J, Xu J, Dang B and Chen G: Inhibition of the NKCC1/NF-κB signaling pathway decreases inflammation and improves brain edema and nerve cell apoptosis in an SBI rat model. Front Mol Neurosci. 14(641993)2021.PubMed/NCBI View Article : Google Scholar | |
Wu MY, Gao F, Tang JF, Shen JC, Gao R, Dang BQ and Chen G: Possible mechanisms of the PERK pathway on neuronal apoptosis in a rat model of surgical brain injury. Am J Transl Res. 13:732–742. 2021.PubMed/NCBI | |
Shen J, Qian M, Wu M, Tang J, Gong Y, Li J, Ji J and Dang B: Rosiglitazone inhibits acyl-CoA synthetase long-chain family number 4 and improves secondary brain injury in a rat model of surgical brain injury. Clin Exp Pharmacol Physiol. 50:927–935. 2023.PubMed/NCBI View Article : Google Scholar | |
Wu M, Gao R, Dang B and Chen G: The blood component iron causes neuronal apoptosis following intracerebral hemorrhage via the PERK pathway. Front Neurol. 11(588548)2020.PubMed/NCBI View Article : Google Scholar | |
Wu M, Wang C, Gong Y, Huang Y, Jiang L, Zhang M, Gao R and Dang B: Potential mechanism of TMEM2/CD44 in endoplasmic reticulum stress-induced neuronal apoptosis in a rat model of traumatic brain injury. Int J Mol Med. 52(119)2023.PubMed/NCBI View Article : Google Scholar | |
Li J, Wu M, Gong Y, Tang J, Shen J, Xu L, Dang B and Chen G: Inhibition of LRRK2-Rab10 pathway improves secondary brain injury after surgical brain injury in rats. Front Surg. 8(749310)2022.PubMed/NCBI View Article : Google Scholar | |
Anthony IC, Ramage SN, Carnie FW, Simmonds P and Bell JE: Does drug abuse alter microglial phenotype and cell turnover in the context of advancing HIV infection? Neuropathol Appl Neurobiol. 31:325–338. 2005.PubMed/NCBI View Article : Google Scholar | |
Haruwaka K, Ikegami A, Tachibana Y, Ohno N, Konishi H, Hashimoto A, Matsumoto M, Kato D, Ono R, Kiyama H, et al: Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat Commun. 10(5816)2019.PubMed/NCBI View Article : Google Scholar | |
Merlini M, Rafalski VA, Ma K, Kim KY, Bushong EA, Rios Coronado PE, Yan Z, Mendiola AS, Sozmen EG, Ryu JK, et al: Microglial Gi-dependent dynamics regulate brain network hyperexcitability. Nat Neurosci. 24:19–23. 2021.PubMed/NCBI View Article : Google Scholar | |
Wang J, Zhang C, Zhu J, Ding J, Chen Y and Han X: Blood-brain barrier disruption and inflammation reaction in mice after chronic exposure to Microcystin-LR. Sci Total Environ. 689:662–678. 2019.PubMed/NCBI View Article : Google Scholar | |
Ueno M, Fujita Y, Tanaka T, Nakamura Y, Kikuta J, Ishii M and Yamashita T: Layer V cortical neurons require microglial support for survival during postnatal development. Nat Neurosci. 16:543–551. 2013.PubMed/NCBI View Article : Google Scholar | |
Corps KN, Roth TL and McGavern DB: Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol. 72:355–362. 2015.PubMed/NCBI View Article : Google Scholar | |
Ikegami A and Wake H: Microglial regulation of blood brain barrier, the neuro-immunological interface. Brain Nerve. 73:913–919. 2021.PubMed/NCBI View Article : Google Scholar : (In Japanese). | |
Planas AM: Role of immune cells migrating to the ischemic brain. Stroke. 49:2261–2267. 2018.PubMed/NCBI View Article : Google Scholar | |
Park GH, Noh H, Shao Z, Ni P, Qin Y, Liu D, Beaudreault CP, Park JS, Abani CP, Park JM, et al: Activated microglia cause metabolic disruptions in developmental cortical interneurons that persist in interneurons from individuals with schizophrenia. Nat Neurosci. 23:1352–1364. 2020.PubMed/NCBI View Article : Google Scholar | |
Zhang Z, Zhang Z, Lu H, Yang Q, Wu H and Wang J: Microglial polarization and inflammatory mediators after intracerebral hemorrhage. Mol Neurobiol. 54:1874–1886. 2017.PubMed/NCBI View Article : Google Scholar | |
Roseborough AD, Zhu Y, Zhao L, Laviolette SR, Pasternak SH and Whitehead SN: Fibrinogen primes the microglial NLRP3 inflammasome and propagates pro-inflammatory signaling via extracellular vesicles: Implications for blood-brain barrier dysfunction. Neurobiol Dis. 177(106001)2023.PubMed/NCBI View Article : Google Scholar | |
Kant R, Halder SK, Fernández JA, Griffin JH and Milner R: Activated protein C attenuates experimental autoimmune encephalomyelitis progression by enhancing vascular integrity and suppressing microglial activation. Front Neurosci. 14(333)2020.PubMed/NCBI View Article : Google Scholar | |
Jolivel V, Bicker F, Binamé F, Ploen R, Keller S, Gollan R, Jurek B, Birkenstock J, Poisa-Beiro L, Bruttger J, et al: Perivascular microglia promote blood vessel disintegration in the ischemic penumbra. Acta Neuropathol. 129:279–295. 2015.PubMed/NCBI View Article : Google Scholar | |
Pan JJ, Qi L, Wang L, Liu C, Song Y, Mamtilahun M, Hu X, Li Y, Chen X, Khan H, et al: M2 microglial extracellular vesicles attenuated blood-brain barrier disruption via MiR-23a-5p in cerebral ischemic mice. Aging Dis. 15:1344–1356. 2024.PubMed/NCBI View Article : Google Scholar | |
Clarner T, Diederichs F, Berger K, Denecke B, Gan L, van der Valk P, Beyer C, Amor S and Kipp M: Myelin debris regulates inflammatory responses in an experimental demyelination animal model and multiple sclerosis lesions. Glia. 60:1468–1480. 2012.PubMed/NCBI View Article : Google Scholar | |
Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA and Stevens B: Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 74:691–705. 2012.PubMed/NCBI View Article : Google Scholar | |
Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, van Wijngaarden P, Wagers AJ, Williams A, Franklin RJM and Ffrench-Constant C: M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci. 16:1211–1218. 2013.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Luo J and Li SY: Nano-curcumin simultaneously protects the blood-brain barrier and reduces M1 microglial activation during cerebral ischemia-reperfusion injury. ACS Appl Mater Interfaces. 11:3763–3770. 2019.PubMed/NCBI View Article : Google Scholar | |
Zhang J, Zheng Y, Luo Y, Du Y, Zhang X and Fu J: Curcumin inhibits LPS-induced neuroinflammation by promoting microglial M2 polarization via TREM2/TLR4/NF-κB pathways in BV2 cells. Mol Immunol. 116:29–37. 2019.PubMed/NCBI View Article : Google Scholar | |
Demirdağ F, Yavuzer S, Cengiz M, Yavuzer H, Kara Z, Ayvacı A, Avcı S, Yürüyen M, Uzun H, Altıparmak MR, et al: The role of NF-κB, PPAR-α, and PPAR-γ in older adults with metabolic syndrome. Horm Metab Res. 55:733–740. 2023.PubMed/NCBI View Article : Google Scholar | |
Li YF, Ren X, Zhang L, Wang YH and Chen T: Microglial polarization in TBI: Signaling pathways and influencing pharmaceuticals. Front Aging Neurosci. 14(901117)2022.PubMed/NCBI View Article : Google Scholar | |
Takuathung MN, Potikanond S, Sookkhee S, Mungkornasawakul P, Jearanaikulvanich T, Chinda K, Wikan N and Nimlamool W: Anti-psoriatic and anti-inflammatory effects of Kaempferia parviflora in keratinocytes and macrophage cells. Biomed Pharmacother. 143(112229)2021.PubMed/NCBI View Article : Google Scholar | |
Ruan Z, Zhang D, Huang R, Sun W, Hou L, Zhao J and Wang Q: Microglial activation damages dopaminergic neurons through MMP-2/-9-mediated increase of blood-brain barrier permeability in a Parkinson's disease mouse model. Int J Mol Sci. 23(2793)2022.PubMed/NCBI View Article : Google Scholar | |
Guo Y, Dai W, Zheng Y, Qiao W, Chen W, Peng L, Zhou H, Zhao T, Liu H, Zheng F and Sun P: Mechanism and regulation of microglia polarization in intracerebral hemorrhage. Molecules. 27(7080)2022.PubMed/NCBI View Article : Google Scholar | |
Tian J, Liu Y, Wang Z, Zhang S, Yang Y, Zhu Y and Yang C: LncRNA Snhg8 attenuates microglial inflammation response and blood-brain barrier damage in ischemic stroke through regulating miR-425-5p mediated SIRT1/NF-κB signaling. J Biochem Mol Toxicol. 35(e22724)2021.PubMed/NCBI View Article : Google Scholar | |
Liao Y, Hu J, Guo C, Wen A, Wen L, Hou Q, Weng Y, Wang J, Ding Y and Yang J: Acteoside alleviates blood-brain barrier damage induced by ischemic stroke through inhibiting microglia HMGB1/TLR4/NLRP3 signaling. Biochem Pharmacol. 220(115968)2024.PubMed/NCBI View Article : Google Scholar | |
Chen G, Hou Y, Li X, Pan R and Zhao D: Sepsis-induced acute lung injury in young rats is relieved by calycosin through inactivating the HMGB1/MyD88/NF-κB pathway and NLRP3 inflammasome. Int Immunopharmacol. 96(107623)2021.PubMed/NCBI View Article : Google Scholar | |
Yin N, Zhao Y, Liu C, Yang Y, Wang ZH, Yu W, Zhang K, Zhang Z, Liu J, Zhang Y and Shi J: Engineered nanoerythrocytes alleviate central nervous system inflammation by regulating the polarization of inflammatory microglia. Adv Mater. 34(e2201322)2022.PubMed/NCBI View Article : Google Scholar | |
Chang H, Ma J, Feng K, Feng N, Wang X, Sun J, Guo T, Wei Y, Xu Y, Wang H, et al: Elevated blood and cerebrospinal fluid biomarkers of microglial activation and blood-brain barrier disruption in anti-NMDA receptor encephalitis. J Neuroinflammation. 20(172)2023.PubMed/NCBI View Article : Google Scholar | |
Yan J, Zhang Y, Wang L, Li Z, Tang S, Wang Y, Gu N, Sun X and Li L: TREM2 activation alleviates neural damage via Akt/CREB/BDNF signalling after traumatic brain injury in mice. J Neuroinflammation. 19(289)2022.PubMed/NCBI View Article : Google Scholar | |
Shi M, Gong Y, Wu M, Gu H, Yu J, Gao F, Ren Z, Qian M, Dang B and Chen G: Downregulation of TREM2/NF-кB signaling may damage the blood-brain barrier and aggravate neuronal apoptosis in experimental rats with surgically injured brain. Brain Res Bull. 183:116–126. 2022.PubMed/NCBI View Article : Google Scholar | |
Sofroniew MV: Astrocyte barriers to neurotoxic inflammation. Nat Rev Neurosci. 16:249–263. 2015.PubMed/NCBI View Article : Google Scholar | |
Cheng X, Wang J, Sun X, Shao L, Guo Z and Li Y: Morphological and functional alterations of astrocytes responding to traumatic brain injury. J Integr Neurosci. 18:203–215. 2019.PubMed/NCBI View Article : Google Scholar | |
Lauranzano E, Rasile M and Matteoli M: Integrating primary astrocytes in a microfluidic model of the blood-brain barrier. Methods Mol Biol. 2492:225–240. 2022.PubMed/NCBI View Article : Google Scholar | |
Yosef N, Xi Y and McCarty JH: Isolation and transcriptional characterization of mouse perivascular astrocytes. PLoS One. 15(e0240035)2020.PubMed/NCBI View Article : Google Scholar | |
Huang J, Ding J, Wang X, Gu C, He Y, Li Y, Fan H, Xie Q, Qi X, Wang Z and Qiu P: Transfer of neuron-derived α-synuclein to astrocytes induces neuroinflammation and blood-brain barrier damage after methamphetamine exposure: Involving the regulation of nuclear receptor-associated protein 1. Brain Behav Immun. 106:247–261. 2022.PubMed/NCBI View Article : Google Scholar | |
Abbott NJ, Rönnbäck L and Hansson E: Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 7:41–53. 2006.PubMed/NCBI View Article : Google Scholar | |
Weber CM, Moiz B, Zic SM, Alpizar Vargas V, Li A and Clyne AM: Induced pluripotent stem cell-derived cells model brain microvascular endothelial cell glucose metabolism. Fluids Barriers CNS. 19(98)2022.PubMed/NCBI View Article : Google Scholar | |
Wu Z, Parry M, Hou XY, Liu MH, Wang H, Cain R, Pei ZF, Chen YC, Guo ZY, Abhijeet S and Chen G: Gene therapy conversion of striatal astrocytes into GABAergic neurons in mouse models of Huntington's disease. Nat Commun. 11(1105)2020.PubMed/NCBI View Article : Google Scholar | |
Bezzi P, Domercq M, Brambilla L, Galli R, Schols D, De Clercq E, Vescovi A, Bagetta G, Kollias G, Meldolesi J and Volterra A: CXCR4-activated astrocyte glutamate release via TNFalpha: Amplification by microglia triggers neurotoxicity. Nat Neurosci. 4:702–710. 2001.PubMed/NCBI View Article : Google Scholar | |
Nedergaard M, Ransom B and Goldman SA: New roles for astrocytes: Redefining the functional architecture of the brain. Trends Neurosci. 26:523–530. 2003.PubMed/NCBI View Article : Google Scholar | |
Simard M and Nedergaard M: The neurobiology of glia in the context of water and ion homeostasis. Neuroscience. 129:877–896. 2004.PubMed/NCBI View Article : Google Scholar | |
Karve IP, Taylor JM and Crack PJ: The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol. 173:692–702. 2016.PubMed/NCBI View Article : Google Scholar | |
Willis EF, MacDonald KPA, Nguyen QH, Garrido AL, Gillespie ER, Harley SBR, Bartlett PF, Schroder WA, Yates AG, Anthony DC, et al: Repopulating microglia promote brain repair in an IL-6-dependent manner. Cell. 180:833–846.e16. 2020.PubMed/NCBI View Article : Google Scholar | |
Lee H and Koh JY: Roles for H+ /K+ -ATPase and zinc transporter 3 in cAMP-mediated lysosomal acidification in bafilomycin A1-treated astrocytes. Glia. 69:1110–1125. 2021.PubMed/NCBI View Article : Google Scholar | |
Chen Y and Swanson RA: Astrocytes and brain injury. J Cereb Blood Flow Metab. 23:137–149. 2003.PubMed/NCBI View Article : Google Scholar | |
Burda JE and Sofroniew MV: Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 81:229–248. 2014.PubMed/NCBI View Article : Google Scholar | |
Lee EJ, Hung YC and Lee MY: Early alterations in cerebral hemodynamics, brain metabolism, and blood-brain barrier permeability in experimental intracerebral hemorrhage. J Neurosurg. 91:1013–1019. 1999.PubMed/NCBI View Article : Google Scholar | |
Lee EJ, Chio CC, Chang CH and Chen HH: Prognostic significance of altered cerebral blood flow velocity in acute head trauma. J Formos Med Assoc. 96:5–12. 1997.PubMed/NCBI | |
Lien CF, Mohanta SK, Frontczak-Baniewicz M, Swinny JD, Zablocka B and Górecki DC: Absence of glial α-dystrobrevin causes abnormalities of the blood-brain barrier and progressive brain edema. J Biol Chem. 287:41374–41385. 2012.PubMed/NCBI View Article : Google Scholar | |
Wolburg H, Noell S, Wolburg-Buchholz K, Mack A and Fallier-Becker P: Agrin, aquaporin-4, and astrocyte polarity as an important feature of the blood-brain barrier. Neuroscientist. 15:180–193. 2009.PubMed/NCBI View Article : Google Scholar | |
Kim I, Moon SO, Park SK, Chae SW and Koh GY: Angiopoietin-1 reduces VEGF-stimulated leukocyte adhesion to endothelial cells by reducing ICAM-1, VCAM-1, and E-selectin expression. Circ Res. 89:477–479. 2001.PubMed/NCBI View Article : Google Scholar | |
Liu B and Neufeld AH: Expression of nitric oxide synthase-2 (NOS-2) in reactive astrocytes of the human glaucomatous optic nerve head. Glia. 30:178–186. 2000.PubMed/NCBI View Article : Google Scholar | |
Jiang L, Pan CL, Wang CY, Liu BQ, Han Y, Hu L, Liu L, Yang Y, Qu JW and Liu WT: Selective suppression of the JNK-MMP2/9 signal pathway by tetramethylpyrazine attenuates neuropathic pain in rats. J Neuroinflammation. 14(174)2017.PubMed/NCBI View Article : Google Scholar | |
Lu L, Hogan-Cann AD, Globa AK, Lu P, Nagy JI, Bamji SX and Anderson CM: Astrocytes drive cortical vasodilatory signaling by activating endothelial NMDA receptors. J Cereb Blood Flow Metab. 39:481–496. 2019.PubMed/NCBI View Article : Google Scholar | |
Mizee MR, Nijland PG, van der Pol SMA, Drexhage JAR, van Het Hof B, Mebius R, van der Valk P, van Horssen J, Reijerkerk A and de Vries HE: Astrocyte-derived retinoic acid: A novel regulator of blood-brain barrier function in multiple sclerosis. Acta Neuropathol. 128:691–703. 2014.PubMed/NCBI View Article : Google Scholar | |
Li Y, Xia Y, Wang Y, Mao L, Gao Y, He Q, Huang M, Chen S and Hu B: Sonic hedgehog (Shh) regulates the expression of angiogenic growth factors in oxygen-glucose-deprived astrocytes by mediating the nuclear receptor NR2F2. Mol Neurobiol. 47:967–975. 2013.PubMed/NCBI View Article : Google Scholar | |
Zacharek A, Chen J, Cui X, Li A, Li Y, Roberts C, Feng Y, Gao Q and Chopp M: Angiopoietin1/Tie2 and VEGF/Flk1 induced by MSC treatment amplifies angiogenesis and vascular stabilization after stroke. J Cereb Blood Flow Metab. 27:1684–1691. 2007.PubMed/NCBI View Article : Google Scholar | |
Chen M, Ba H, Lu C, Dai J and Sun J: Glial cell line-derived neurotrophic factor (GDNF) promotes angiogenesis through the demethylation of the fibromodulin (FMOD) promoter in glioblastoma. Med Sci Monit. 24:6137–6143. 2018.PubMed/NCBI View Article : Google Scholar | |
Rodriguez-Perez AI, Borrajo A, Diaz-Ruiz C, Garrido-Gil P and Labandeira-Garcia JL: Crosstalk between insulin-like growth factor-1 and angiotensin-II in dopaminergic neurons and glial cells: Role in neuroinflammation and aging. Oncotarget. 7:30049–30067. 2016.PubMed/NCBI View Article : Google Scholar | |
Cao F, Jiang Y, Wu Y, Zhong J, Liu J, Qin X, Chen L, Vitek MP, Li F, Xu L and Sun X: Apolipoprotein E-mimetic COG1410 reduces acute vasogenic edema following traumatic brain injury. J Neurotrauma. 33:175–182. 2016.PubMed/NCBI View Article : Google Scholar | |
Leybaert L: Neurobarrier coupling in the brain: A partner of neurovascular and neurometabolic coupling? J Cereb Blood Flow Metab. 25:2–16. 2005.PubMed/NCBI View Article : Google Scholar | |
Chen Z, Kelly JR, Morales JE, Sun RC, De A, Burkin DJ and McCarty JH: The alpha7 integrin subunit in astrocytes promotes endothelial blood-brain barrier integrity. Development. 150(dev201356)2023.PubMed/NCBI View Article : Google Scholar | |
Mayer U, Saher GFässler R, Bornemann A, Echtermeyer F, von der Mark H, Miosge N, Pöschl E and von der Mark K: Absence of integrin alpha 7 causes a novel form of muscular dystrophy. Nat Genet. 17:318–323. 1997.PubMed/NCBI View Article : Google Scholar | |
Mielenz D, Hapke S, Pöschl E, von Der Mark H and von Der Mark K: The integrin alpha 7 cytoplasmic domain regulates cell migration, lamellipodia formation, and p130CAS/Crk coupling. J Biol Chem. 276:13417–13426. 2001.PubMed/NCBI View Article : Google Scholar | |
Kim H, Leng K, Park J, Sorets AG, Kim S, Shostak A, Embalabala RJ, Mlouk K, Katdare KA, Rose IVL, et al: Reactive astrocytes transduce inflammation in a blood-brain barrier model through a TNF-STAT3 signaling axis and secretion of alpha 1-antichymotrypsin. Nat Commun. 13(6581)2022.PubMed/NCBI View Article : Google Scholar | |
Lee LL, Aung HH, Wilson DW, Anderson SE, Rutledge JC and Rutkowsky JM: Triglyceride-rich lipoprotein lipolysis products increase blood-brain barrier transfer coefficient and induce astrocyte lipid droplets and cell stress. Am J Physiol Cell Physiol. 312:C500–C516. 2017.PubMed/NCBI View Article : Google Scholar | |
Malik VA, Zajicek F, Mittmann LA, Klaus J, Unterseer S, Rajkumar S, Pütz B, Deussing JM, Neumann ID, Rupprecht R and Di Benedetto B: GDF15 promotes simultaneous astrocyte remodeling and tight junction strengthening at the blood-brain barrier. J Neurosci Res. 98:1433–1456. 2020.PubMed/NCBI View Article : Google Scholar | |
Shimizu F, Sano Y, Tominaga O, Maeda T, Abe MA and Kanda T: Advanced glycation end-products disrupt the blood-brain barrier by stimulating the release of transforming growth factor-β by pericytes and vascular endothelial growth factor and matrix metalloproteinase-2 by endothelial cells in vitro. Neurobiol Aging. 34:1902–1912. 2013.PubMed/NCBI View Article : Google Scholar | |
Senatorov VV Jr, Friedman AR, Milikovsky DZ, Ofer J, Saar-Ashkenazy R, Charbash A, Jahan N, Chin G, Mihaly E, Lin JM, et al: Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction. Sci Transl Med. 11(eaaw8283)2019.PubMed/NCBI View Article : Google Scholar | |
Guo Y, Dong L, Gong A, Zhang J, Jing L, Ding T, Li PA and Zhang JZ: Damage to the blood-brain barrier and activation of neuroinflammation by focal cerebral ischemia under hyperglycemic condition. Int J Mol Med. 48(142)2021.PubMed/NCBI View Article : Google Scholar | |
Wang QS, Ding HG, Chen SL, Liu XQ, Deng YY, Jiang WQ, Li Y, Huang LQ, Han YL, Wen MY, et al: Hypertonic saline mediates the NLRP3/IL-1β signaling axis in microglia to alleviate ischemic blood-brain barrier permeability by downregulating astrocyte-derived VEGF in rats. CNS Neurosci Ther. 26:1045–1057. 2020.PubMed/NCBI View Article : Google Scholar | |
You L, Yu PP, Dong T, Guo W, Chang S, Zheng B, Ci Y, Wang F, Yu P, Gao G and Chang YZ: Astrocyte-derived hepcidin controls iron traffic at the blood-brain-barrier via regulating ferroportin 1 of microvascular endothelial cells. Cell Death Dis. 13(667)2022.PubMed/NCBI View Article : Google Scholar | |
Pasti L, Volterra A, Pozzan T and Carmignoto G: Intracellular calcium oscillations in astrocytes: A highly plastic, bidirectional form of communication between neurons and astrocytes in situ. J Neurosci. 17:7817–7830. 1997.PubMed/NCBI View Article : Google Scholar | |
Huber JD, Egleton RD and Davis TP: Molecular physiology and pathophysiology of tight junctions in the blood-brain barrier. Trends Neurosci. 24:719–725. 2001.PubMed/NCBI View Article : Google Scholar | |
Cotrina ML, Lin JH, Alves-Rodrigues A, Liu S, Li J, Azmi-Ghadimi H, Kang J, Naus CC and Nedergaard M: Connexins regulate calcium signaling by controlling ATP release. Proc Natl Acad Sci USA. 95:15735–15740. 1998.PubMed/NCBI View Article : Google Scholar | |
Sneyd J, Charles AC and Sanderson MJ: A model for the propagation of intercellular calcium waves. Am J Physiol. 266:C293–C302. 1994.PubMed/NCBI View Article : Google Scholar | |
Chapouly C, Tadesse Argaw A, Horng S, Castro K, Zhang J, Asp L, Loo H, Laitman BM, Mariani JN, Straus Farber R, et al: Astrocytic TYMP and VEGFA drive blood-brain barrier opening in inflammatory central nervous system lesions. Brain. 138:1548–1567. 2015.PubMed/NCBI View Article : Google Scholar | |
Chu H, Yang X, Huang C, Gao Z, Tang Y and Dong Q: Apelin-13 protects against ischemic blood-brain barrier damage through the effects of aquaporin-4. Cerebrovasc Dis. 44:10–25. 2017.PubMed/NCBI View Article : Google Scholar | |
Jackson RJ, Meltzer JC, Nguyen H, Commins C, Bennett RE, Hudry E and Hyman BT: APOE4 derived from astrocytes leads to blood-brain barrier impairment. Brain. 145:3582–3593. 2022.PubMed/NCBI View Article : Google Scholar | |
Qin X, Wang J, Chen S, Liu G, Wu C, Lv Q, He X, Bai X, Huang W and Liao H: Astrocytic p75NTR expression provoked by ischemic stroke exacerbates the blood-brain barrier disruption. Glia. 70:892–912. 2022.PubMed/NCBI View Article : Google Scholar | |
Lee SW, Kim WJ, Choi YK, Song HS, Son MJ, Gelman IH, Kim YJ and Kim KW: SSeCKS regulates angiogenesis and tight junction formation in blood-brain barrier. Nat Med. 9:900–906. 2003.PubMed/NCBI View Article : Google Scholar | |
Takarada-Iemata M, Yoshikawa A, Ta HM, Okitani N, Nishiuchi T, Aida Y, Kamide T, Hattori T, Ishii H, Tamatani T, et al: N-myc downstream-regulated gene 2 protects blood-brain barrier integrity following cerebral ischemia. Glia. 66:1432–1446. 2018.PubMed/NCBI View Article : Google Scholar | |
Tian W, Sawyer A, Kocaoglu FB and Kyriakides TR: Astrocyte-derived thrombospondin-2 is critical for the repair of the blood-brain barrier. Am J Pathol. 179:860–868. 2011.PubMed/NCBI View Article : Google Scholar | |
Jayakumar AR, Tong XY, Ruiz-Cordero R, Bregy A, Bethea JR, Bramlett HM and Norenberg MD: Activation of NF-κB mediates astrocyte swelling and brain edema in traumatic brain injury. J Neurotrauma. 31:1249–1257. 2014.PubMed/NCBI View Article : Google Scholar | |
Wu M, Gong Y, Jiang L, Zhang M, Gu H, Shen H and Dang B: VEGF regulates the blood-brain barrier through MMP-9 in a rat model of traumatic brain injury. Exp Ther Med. 24(728)2022.PubMed/NCBI View Article : Google Scholar | |
Wu MY, Gao F, Yang XM, Qin X, Chen GZ, Li D, Dang BQ and Chen G: Matrix metalloproteinase-9 regulates the blood brain barrier via the hedgehog pathway in a rat model of traumatic brain injury. Brain Res. 1727(146553)2020.PubMed/NCBI View Article : Google Scholar | |
Persidsky Y, Ramirez SH, Haorah J and Kanmogne GD: Blood-brain barrier: Structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol. 1:223–236. 2006.PubMed/NCBI View Article : Google Scholar | |
Kassan M, Kwon Y, Munkhsaikhan U, Sahyoun AM, Ishrat T, Galán M, Gonzalez AA, Abidi AH, Kassan A and Ait-Aissa K: Protective role of short-chain fatty acids against Ang-II-induced mitochondrial dysfunction in brain endothelial cells: A potential role of heme oxygenase 2. Antioxidants (Basel). 12(160)2023.PubMed/NCBI View Article : Google Scholar | |
Wang YI, Abaci HE and Shuler ML: Microfluidic blood-brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng. 114:184–194. 2017.PubMed/NCBI View Article : Google Scholar | |
Min XL, Zou H, Yan J, Lyu Q, He X and Shang FF: Stress conditions induced circRNAs profile of extracellular vesicles in brain microvascular endothelial cells. Metab Brain Dis. 37:1977–1987. 2022.PubMed/NCBI View Article : Google Scholar | |
Furtado D, Björnmalm M, Ayton S, Bush AI, Kempe K and Caruso F: Overcoming the blood-brain barrier: The role of nanomaterials in treating neurological diseases. Adv Mater. 30(e1801362)2018.PubMed/NCBI View Article : Google Scholar | |
Grammas P, Martinez J and Miller B: Cerebral microvascular endothelium and the pathogenesis of neurodegenerative diseases. Expert Rev Mol Med. 13(e19)2011.PubMed/NCBI View Article : Google Scholar | |
Oldendorf WH, Cornford ME and Brown WJ: The large apparent work capability of the blood-brain barrier: A study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat. Ann Neurol. 1:409–417. 1977.PubMed/NCBI View Article : Google Scholar | |
Grutzendler J and Nedergaard M: Cellular control of brain capillary blood flow: In vivo imaging veritas. Trends Neurosci. 42:528–536. 2019.PubMed/NCBI View Article : Google Scholar | |
Raut S, Patel R and Al-Ahmad AJ: Presence of a mutation in PSEN1 or PSEN2 gene is associated with an impaired brain endothelial cell phenotype in vitro. Fluids Barriers CNS. 18(3)2021.PubMed/NCBI View Article : Google Scholar | |
Lisk C, McCord J, Bose S, Sullivan T, Loomis Z, Nozik-Grayck E, Schroeder T, Hamilton K and Irwin DC: Nrf2 activation: A potential strategy for the prevention of acute mountain sickness. Free Radic Biol Med. 63:264–273. 2013.PubMed/NCBI View Article : Google Scholar | |
Nicolicht-Amorim P, Delgado-Garcia LM, Nakamura TKE, Courbassier NR, Mosini AC and Porcionatto MA: Simple and efficient protocol to isolate and culture brain microvascular endothelial cells from newborn mice. Front Cell Neurosci. 16(949412)2022.PubMed/NCBI View Article : Google Scholar | |
Sawada N: Tight junction-related human diseases. Pathol Int. 63:1–12. 2013.PubMed/NCBI View Article : Google Scholar | |
González-Mariscal L, Posadas Y, Miranda J, Uc PY, Ortega-Olvera JM and Hernández S: Strategies that target tight junctions for enhanced drug delivery. Curr Pharm Des. 22:5313–5346. 2016.PubMed/NCBI View Article : Google Scholar | |
Keaney J and Campbell M: The dynamic blood-brain barrier. FEBS J. 282:4067–4079. 2015.PubMed/NCBI View Article : Google Scholar | |
Kumar R, Sharma A and Tiwari RK: Can we predict blood brain barrier permeability of ligands using computational approaches? Interdiscip Sci. 5:95–101. 2013.PubMed/NCBI View Article : Google Scholar | |
Saxena D, Sharma A, Siddiqui MH and Kumar R: Blood brain barrier permeability prediction using machine learning techniques: An update. Curr Pharm Biotechnol. 20:1163–1171. 2019.PubMed/NCBI View Article : Google Scholar | |
Song D, Jiang X, Liu Y, Sun Y, Cao S and Zhang Z: Asiaticoside attenuates cell growth inhibition and apoptosis induced by Aβ1-42 via inhibiting the TLR4/NF-κB signaling pathway in human brain microvascular endothelial cells. Front Pharmacol. 9(28)2018.PubMed/NCBI View Article : Google Scholar | |
Uraoka M, Ikeda K, Kurimoto-Nakano R, Nakagawa Y, Koide M, Akakabe Y, Kitamura Y, Ueyama T, Matoba S, Yamada H, et al: Loss of bcl-2 during the senescence exacerbates the impaired angiogenic functions in endothelial cells by deteriorating the mitochondrial redox state. Hypertension. 58:254–263. 2011.PubMed/NCBI View Article : Google Scholar | |
Rajeev V, Fann DY, Dinh QN, Kim HA, De Silva TM, Lai MKP, Chen CL, Drummond GR, Sobey CG and Arumugam TV: Pathophysiology of blood brain barrier dysfunction during chronic cerebral hypoperfusion in vascular cognitive impairment. Theranostics. 12:1639–1658. 2022.PubMed/NCBI View Article : Google Scholar | |
Sweeney MD, Sagare AP and Zlokovic BV: Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 14:133–150. 2018.PubMed/NCBI View Article : Google Scholar | |
Bernard-Patrzynski F, Lécuyer MA, Puscas I, Boukhatem I, Charabati M, Bourbonnière L, Ramassamy C, Leclair G, Prat A and Roullin VG: Isolation of endothelial cells, pericytes and astrocytes from mouse brain. PLoS One. 14(e0226302)2019.PubMed/NCBI View Article : Google Scholar | |
Vajtr D, Benada O, Kukacka J, Průša R, Houstava L, Ťoupalík P and Kizek R: Correlation of ultrastructural changes of endothelial cells and astrocytes occurring during blood brain barrier damage after traumatic brain injury with biochemical markers of BBB leakage and inflammatory response. Physiol Res. 58:263–268. 2009.PubMed/NCBI View Article : Google Scholar | |
Yao X, Uchida K, Papadopoulos MC, Zador Z, Manley GT and Verkman AS: Mildly reduced brain swelling and improved neurological outcome in aquaporin-4 knockout mice following controlled cortical impact brain injury. J Neurotrauma. 32:1458–1464. 2015.PubMed/NCBI View Article : Google Scholar | |
Abbott NJ: Astrocyte-endothelial interactions and blood-brain barrier permeability. J Anat. 200:629–638. 2002.PubMed/NCBI View Article : Google Scholar | |
Liu H, Wei JY, Li Y, Ban M, Sun Q, Wang HJ, Zhao D, Tong PG, Wang L, Wang KJ, et al: Endothelial depletion of Atg7 triggers astrocyte-microvascular disassociation at blood-brain barrier. J Cell Biol. 222(e202103098)2023.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Wu J, Wang J, He L, Lai H, Zhang T, Wang X and Li W: Mitochondrial oxidative stress in brain microvascular endothelial cells: Triggering blood-brain barrier disruption. Mitochondrion. 69:71–82. 2023.PubMed/NCBI View Article : Google Scholar | |
Sun P, Zhang K, Hassan SH, Zhang X, Tang X, Pu H, Stetler RA, Chen J and Yin KJ: Endothelium-targeted deletion of microRNA-15a/16-1 promotes poststroke angiogenesis and improves long-term neurological recovery. Circ Res. 126:1040–1057. 2020.PubMed/NCBI View Article : Google Scholar | |
Ren C, Li N, Li S, Han R, Huang Q, Hu J, Jin K and Ji X: Limb ischemic conditioning improved cognitive deficits via eNOS-dependent augmentation of angiogenesis after chronic cerebral hypoperfusion in rats. Aging Dis. 9:869–879. 2018.PubMed/NCBI View Article : Google Scholar | |
Zhu HY, Hong FF and Yang SL: The roles of nitric oxide synthase/nitric oxide pathway in the pathology of vascular dementia and related therapeutic approaches. Int J Mol Sci. 22(4540)2021.PubMed/NCBI View Article : Google Scholar | |
Ungvari Z, Tarantini S, Kiss T, Wren JD, Giles CB, Griffin CT, Murfee WL, Pacher P and Csiszar A: Endothelial dysfunction and angiogenesis impairment in the ageing vasculature. Nat Rev Cardiol. 15:555–565. 2018.PubMed/NCBI View Article : Google Scholar | |
Hübner K, Cabochette P, Diéguez-Hurtado R, Wiesner C, Wakayama Y, Grassme KS, Hubert M, Guenther S, Belting HG, Affolter M, et al: Wnt/β-catenin signaling regulates VE-cadherin-mediated anastomosis of brain capillaries by counteracting S1pr1 signaling. Nat Commun. 9(4860)2018.PubMed/NCBI View Article : Google Scholar | |
Swaminathan B, Youn SW, Naiche LA, Du J, Villa SR, Metz JB, Feng H, Zhang C, Kopan R, Sims PA and Kitajewski JK: Endothelial Notch signaling directly regulates the small GTPase RND1 to facilitate Notch suppression of endothelial migration. Sci Rep. 12(1655)2022.PubMed/NCBI View Article : Google Scholar | |
Zhu T, Xie WJ, Wang L, Jin XB, Meng XB, Sun GB and Sun XB: Notoginsenoside R1 activates the NAMPT-NAD+-SIRT1 cascade to promote postischemic angiogenesis by modulating Notch signaling. Biomed Pharmacother. 140(111693)2021.PubMed/NCBI View Article : Google Scholar | |
Ma C, Zhou J, Xu X, Wang L, Qin S, Hu C, Nie L and Tu Y: The construction of a radiation-induced brain injury model and preliminary study on the effect of human recombinant endostatin in treating radiation-induced brain injury. Med Sci Monit. 25:9392–9401. 2019.PubMed/NCBI View Article : Google Scholar | |
Deng Z, Zhou L, Wang Y, Liao S, Huang Y, Shan Y, Tan S, Zeng Q, Peng L, Huang H and Lu Z: Astrocyte-derived VEGF increases cerebral microvascular permeability under high salt conditions. Aging (Albany NY). 12:11781–11793. 2020.PubMed/NCBI View Article : Google Scholar | |
Lee WH, Warrington JP, Sonntag WE and Lee YW: Irradiation alters MMP-2/TIMP-2 system and collagen type IV degradation in brain. Int J Radiat Oncol Biol Phys. 82:1559–1566. 2012.PubMed/NCBI View Article : Google Scholar | |
Kisler K, Nelson AR, Rege SV, Ramanathan A, Wang Y, Ahuja A, Lazic D, Tsai PS, Zhao Z, Zhou Y, et al: Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain. Nat Neurosci. 20:406–416. 2017.PubMed/NCBI View Article : Google Scholar | |
Kur J, Newman EA and Chan-Ling T: Cellular and physiological mechanisms underlying blood flow regulation in the retina and choroid in health and disease. Prog Retin Eye Res. 31:377–406. 2012.PubMed/NCBI View Article : Google Scholar | |
Mae MA, He L, Nordling S, Vazquez-Liebanas E, Nahar K, Jung B, Li X, Tan BC, Chin Foo J, Cazenave-Gassiot A, et al: Single-cell analysis of blood-brain barrier response to pericyte loss. Circ Res. 128:e46–e62. 2021.PubMed/NCBI View Article : Google Scholar | |
Prager O, Kamintsky L, Hasam-Henderson LA, Schoknecht K, Wuntke V, Papageorgiou I, Swolinsky J, Muoio V, Bar-Klein G, Vazana U, et al: Seizure-induced microvascular injury is associated with impaired neurovascular coupling and blood-brain barrier dysfunction. Epilepsia. 60:322–336. 2019.PubMed/NCBI View Article : Google Scholar | |
Stefanska A, Kenyon C, Christian HC, Buckley C, Shaw I, Mullins JJ and Péault B: Human kidney pericytes produce renin. Kidney Int. 90:1251–1261. 2016.PubMed/NCBI View Article : Google Scholar | |
Korte N, James G, You H, Hirunpattarasilp C, Christie I, Sethi H and Attwell D: Noradrenaline released from locus coeruleus axons contracts cerebral capillary pericytes via α2 adrenergic receptors. J Cereb Blood Flow Metab. 43:1142–1152. 2023.PubMed/NCBI View Article : Google Scholar | |
Huang H: Pericyte-endothelial interactions in the retinal microvasculature. Int J Mol Sci. 21(7413)2020.PubMed/NCBI View Article : Google Scholar | |
Dehouck MP, Tachikawa M, Hoshi Y, Omori K, Maurage CA, Strecker G, Dehouck L, Boucau MC, Uchida Y, Gosselet F, et al: Quantitative targeted absolute proteomics for better characterization of an in vitro human blood-brain barrier model derived from hematopoietic stem cells. Cells. 11(3963)2022.PubMed/NCBI View Article : Google Scholar | |
Winkler EA, Sengillo JD, Bell RD, Wang J and Zlokovic BV: Blood-spinal cord barrier pericyte reductions contribute to increased capillary permeability. J Cereb Blood Flow Metab. 32:1841–1852. 2012.PubMed/NCBI View Article : Google Scholar | |
Armulik A, Genové G and Betsholtz C: Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 21:193–215. 2011.PubMed/NCBI View Article : Google Scholar | |
Figueira I, Galego S, Custódio-Santos T, Vicente R, Molnár K, Haskó J, Malhó R, Videira M, Wilhelm I, Krizbai I and Brito MA: Picturing breast cancer brain metastasis development to unravel molecular players and cellular crosstalk. Cancers (Basel). 13(910)2021.PubMed/NCBI View Article : Google Scholar | |
Alarcon-Martinez L, Villafranca-Baughman D, Quintero H, Kacerovsky JB, Dotigny F, Murai KK, Prat A, Drapeau P and Di Polo A: Interpericyte tunnelling nanotubes regulate neurovascular coupling. Nature. 585:91–95. 2020.PubMed/NCBI View Article : Google Scholar | |
Lechertier T, Reynolds LE, Kim H, Pedrosa AR, Gómez-Escudero J, Muñoz-Félix JM, Batista S, Dukinfield M, Demircioglu F, Wong PP, et al: Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth. Nat Commun. 11(2810)2020.PubMed/NCBI View Article : Google Scholar | |
Wu Y, Fu J, Huang Y, Duan R, Zhang W, Wang C, Wang S, Hu X, Zhao H, Wang L, et al: Biology and function of pericytes in the vascular microcirculation. Animal Model Exp Med. 6:337–345. 2023.PubMed/NCBI View Article : Google Scholar | |
Daneman R, Zhou L, Kebede AA and Barres BA: Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature. 468:562–566. 2010.PubMed/NCBI View Article : Google Scholar | |
Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R and Zlokovic BV: Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron. 68:409–427. 2010.PubMed/NCBI View Article : Google Scholar | |
Abramsson A, Kurup S, Busse M, Yamada S, Lindblom P, Schallmeiner E, Stenzel D, Sauvaget D, Ledin J, Ringvall M, et al: Defective N-sulfation of heparan sulfate proteoglycans limits PDGF-BB binding and pericyte recruitment in vascular development. Genes Dev. 21:316–331. 2007.PubMed/NCBI View Article : Google Scholar | |
Weidenfeller C, Svendsen CN and Shusta EV: Differentiating embryonic neural progenitor cells induce blood-brain barrier properties. J Neurochem. 101:555–565. 2007.PubMed/NCBI View Article : Google Scholar | |
Huang Y, Chen S, Luo Y and Han Z: Crosstalk between inflammation and the BBB in stroke. Curr Neuropharmacol. 18:1227–1236. 2020.PubMed/NCBI View Article : Google Scholar | |
Thurgur H and Pinteaux E: Microglia in the neurovascular unit: Blood-brain barrier-microglia interactions after central nervous system disorders. Neuroscience. 405:55–67. 2019.PubMed/NCBI View Article : Google Scholar | |
Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, He L, Norlin J, Lindblom P, Strittmatter K, et al: Pericytes regulate the blood-brain barrier. Nature. 468:557–561. 2010.PubMed/NCBI View Article : Google Scholar | |
Hellström M, Kalén M, Lindahl P, Abramsson A and Betsholtz C: Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development. 126:3047–3055. 1999.PubMed/NCBI View Article : Google Scholar | |
Li J, Li M, Ge Y, Chen J, Ma J, Wang C, Sun M, Wang L, Yao S and Yao C: β-amyloid protein induces mitophagy-dependent ferroptosis through the CD36/PINK/PARKIN pathway leading to blood-brain barrier destruction in Alzheimer's disease. Cell Biosci. 12(69)2022.PubMed/NCBI View Article : Google Scholar | |
Ma Q, Zhao Z, Sagare AP, Wu Y, Wang M, Owens NC, Verghese PB, Herz J, Holtzman DM and Zlokovic BV: Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism. Mol Neurodegener. 13(57)2018.PubMed/NCBI View Article : Google Scholar | |
Bongarzone S, Savickas V, Luzi F and Gee AD: Targeting the receptor for advanced glycation endproducts (RAGE): A medicinal chemistry perspective. J Med Chem. 60:7213–7232. 2017.PubMed/NCBI View Article : Google Scholar | |
Bell-Temin H, Culver-Cochran AE, Chaput D, Carlson CM, Kuehl M, Burkhardt BR, Bickford PC, Liu B and Stevens SM Jr: Novel molecular insights into classical and alternative activation states of microglia as revealed by stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics. Mol Cell Proteomics. 14:3173–3184. 2015.PubMed/NCBI View Article : Google Scholar | |
Becerra-Calixto A and Cardona-Gómez GP: The role of astrocytes in neuroprotection after brain stroke: Potential in cell therapy. Front Mol Neurosci. 10(88)2017.PubMed/NCBI View Article : Google Scholar |