Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Oct 1.
Published in final edited form as: Biochim Biophys Acta. 2009 Oct 29;1802(10):881–888. doi: 10.1016/j.bbadis.2009.10.011

Mouse Models of Neurological Disorders: A View From the Blood-brain Barrier

William A Banks 1
PMCID: PMC2891624  NIHMSID: NIHMS156361  PMID: 19879356

Abstract

The number of disease models that involve an aspect of blood-brain barrier (BBB) dysregulation have increased tremendously. The main factors contributing to this expansion have been an increased number of diseases in which the BBB is known to be involved, an increase in the known functions of the BBB, and an increase in the number of models and tools with which those diverse functions can be studied. In many cases, the BBB may be a target of disease; current thinking would include hypertensive encephalopathy and perhaps stroke in this category. Another category are those diseases in which special attributes of the BBB may predispose to disease; for example, the ability of a pathogen to cross the BBB often depends on the pathogen’s ability to invoke trancytotic pathways in the brain endothelial or choroid plexus cell. Of special interest are those diseases in which the BBB may be the primary seat of disease or play a major role in the onset or progression of the disease. An increasing number of diseases are so categorized in which BBB dysfunction or dysregulation plays a major role; this review highlights such roles for the BBB including those proposed for Alzheimer’s disease and obesity.

Keywords: Blood-brain Barrier, Models; Stroke; Obesity; Alzheimer’s Disease; Multiple Sclerosis; Transcytosis; Paracellular; Neuroimmune; Choroid Plexus

Introduction

Models of blood-brain barrier (BBB)-disease interactions have increased tremendously in number in the last two decades. Although there are many reasons for this, three main trends are largely responsible for the increase. First, there are more diseases which are thought to involve the BBB. At the same time, the explosion in animal models for any given disease have further enhanced this reason. Second, there are more better and models for studying the BBB. This allows functions to be investigated that previously could not be modeled. Third, the BBB has more functions than were previously appreciated. These functions can be studied and their dysfunction can participate in disease processes in ways which were not imagined even a few years ago. These three aspects are considered in more detail below.

The trends and future directions can be better appreciated by examining the state of BBB models 3–4 decades ago. Good sources for this are Rapoport’s classic “Blood-Brain Barrier in Physiology and Medicine” published in 1976 [1]and Bradbury’s “The Concept of a Blood-Brain Barrier”published in 1979[2]. These books remain excellent sources for understanding the fundamentals of the BBB, but also provide snap shots of the status of BBB disease models some 30+ years ago. Many things have not changed: models of multiple sclerosis and stroke are still of major interest, debate continues about whether immune cells cross the BBB by penetrating between or through endothelial cells, and issues related to drug delivery to the CNS present major challenges. But many things have changed also: there are many more models of stroke as well as models of components of stroke (such as hypoxia/reoxygenation), immune cells are now widely believed to cross the BBB under normal physiologic conditions and not just during brain infection, and drug delivery has become still more challenging with the discovery of, for example, efflux systems. Diseases most actively modeled in the 1970’s were stroke, multiple sclerosis, trauma, cytotoxicity, invasion by cancer cells and leukemias, brain infections and encephalitides, convulsions and epilepsy, and hypertensive encephalopathy. BBB tissues were known to be enzymatically active and to be important in preventing monoamine transport across the BBB. Methods available allowed three generic types of studies to be done: the measure of uptake rates of substances which crossed the BBB very rapidly, such as hexoses, amino acids, or small lipid soluble molecules; the study of fluxes of substances which were metabolically stable such as electrolytes; studies of BBB disruption and edema, often using dye or fluid shift models. The conceptual view of the functions of the BBB these models helped to define and exploit were that of the nutritional and homeostatic regulatory interface, the lipid permeable cell membrane, and the barrier.

General Themes

The BBB models of diseases can be subdivided into 3 general categories as to their relation to the disease that they model. The first and traditionally the most common category is that of inducing some aspect of the disease by a mechanism that is clearly not the etiology of the disease. The purpose of these studies is to investigate the downstream consequences of the disease feature while recognizing that upstream events cannot be studied. An example of this model is induction of type I diabetes mellitus with streptozotocin. Although streptozotocin induces an insulinopenic hyperglycemic state, the hallmark of type I diabetes mellitus, streptozotocin is not the common etiology of diabetes mellitus in humans. As a result, the downstream consequences of deficient insulin or hyperglycemia can be studied, but questions related to etiology, predisposition to disease, or genetics are not easily studied with this model.

A second category is that in which the disease being studied arises from essentially the same etiology in the model as in the human and follows essentially the same course. Pure examples of these are rare and the most useful models. An example in this category is that of maturity onset obesity in a mouse strain which is not otherwise prone to obesity. In a version of this model, randomly selected mice are fed regular chow for an extended period. A subset will tend towards a higher level of obesity than the average, just as in the human population. In a variation of this version, a high fat diet and sedentary conditions will accentuate the differences in body weight, just as occurs in humans.

A third category of model is that in which the relation between the etiology of the disease and the inducer of the model are unclear. This leads to ambiguity as to what extent the model recapitulates the disease. An example of this is the use of experimental allergic encephalomyelitis (EAE) to model multiple sclerosis. There are several EAE models that primarily vary in the method used to induce disease, but most activate the immune system against components of the nervous system. As the etiology of multiple sclerosis is unknown, it is unclear to what degree, if any, the various methods used to induce EAE resemble the etiology of multiple sclerosis.

Diseases Affecting or Involving the BBB

The number of diseases in which the BBB is thought to be affected has increased dramatically. Included are some of the most important diseases of Western society such as Alzheimer’s disease, obesity, epilepsy, and diabetes mellitus [3-8]. The role of the BBB in many diseases has also shifted from being a passive target to an active participant, even a prime participant, in disease. Two examples of diseases in which the BBB may play the role of prime participant are obesity and Alzheimer’s disease; these are considered in detail below.

Other models suggesting a primary role for the BBB in disease states are those relating to immune cell and pathogen penetration of the BBB. Diseases involving immune infiltration are often mistakenly thought to occur because a disruption of the BBB allows the immune cells to leak into the brain. It is now clearly understood that immune cells and brain endothelial cells participate in an elaborate cross talk termed diapedesis [9]. Passage of immune cells across the BBB involves activation of both the immune cell and BBB cells [10;11]. Blockade of key receptors on either brain endothelial cells or the immune cells can halt or retard diapedesis. This is most dramatically shown by treatment with natalizumab, an antibody that binds to immune cells to block alpha(4) integrin binding and is effective against multiple sclerosis (3206}.

Multiple sclerosis is a disease in which the BBB plays a major role. EAE is used as a model of multiple sclerosis [12]. There are several versions of induction of EAE that produce either relapsing and non-relapsing disease [13;14]. EAE models clearly support a role of the BBB in mediating onset and progression of disease [15;16]. The EAE models have also been important tools in the investigation of how immune cells cross the BBB [17]. Work with these models also shows that disruption of the BBB is secondary to immune cell invasion, not the other way round [18-20]. Other aspects of BBB function are also likely affected by induction of EAE and, presumably, in multiple sclerosis such as cytokine transport [21]. The EAE model has also served as an example of how neuroimmune activation can facilitate immune cell invasion for other conditions and diseases such as spinal cord injury [22].

HIV-1 invasion of the CNS occurs when immune cells infected with the virus cross the BBB [23]. Immune cells containing HIV-1 are activated and more likely to adhere to brain endothelial cells than non-infected cells [24]. Models of this disease also show that immune cell invasion produces BBB leakage and disruption rather than the BBB disruption allowing immune cells to leak into the brain [25]. HIV-1 can also cross the BBB as free virus and does so by interacting in a complex manner with the cells which constitute the BBB [26;27]. This complexity occurs not just with initial binding, but with subsequent interaction with cellular machinery that controls transcytosis [28-31] Indeed, the neuroAIDS field has produced an important example of how pathogen and BBB cells interact to activate one another and to forward the progression of disease [24;32]. This interaction has consequences in addition to the facilitation of passage of pathogen across the BBB [23;33]. For example, the immune activation induced by the neuroAIDS process [34] activates other components of the neurovascular unit, secretions from BBB cells, and alters expression of p-glycoprotein at the BBB [35-37]. This latter effect, in turn, affects the delivery of antivirals to the CNS [38;39].

These principles apply to most other neuropathogens which enter the CNS by crossing the vascular BBB or choroid plexus [40]. That is, the virus, bacteria, or parasite induces alterations in the BBB function which facilitate the passage of the agent across the barrier as opposed to leaking across an already disrupted BBB [41-44]. The agent often induces cooperation of the BBB, taking over some aspect of endogenous machinery.

Models of the BBB

The multiplication of models can be illustrated by the increased number of models for stroke or diabetes mellitus. Stroke models or models relating to some aspect of stroke have expanded greatly in the last few decades and demonstrated important changes in BBB tight junctions in the post-ischemic state [45]. For example, there are currently five major categories of ischemic models: embolic middle cerebral artery occlusion, endovascular filament middle cerebral artery occlusion, permanant transcranial midddle cerebral artery occlsion, transient transcranial middle cerebral artery occluson, and cerebrocortical photothrombosis. These models are not simply variants on how to produce the same end point, but have subtle differences including how the BBB is affected or responds. There are also “component models” that involve replication of a major aspect of stroke [46]. For example, hypoxia/reoxygenation models lower the ambient oxygen to a level and for a time that affects BBB and CNS function to study the effects of hypoxia. Oxygen levels are then returned to normal to study effects of reoxygenation, a period when damage from oxidative stress can occur. In this model, unlike the focal ischemia models, blood flow does not cease, no physical damage is done to endothelial cells by a plug or clot, and hypoxia/reoxygenation is global, not localized. As stroke involves all of these, hypoxia/reoxygenation is not a stroke model, although it models a major component of stroke. Results may be applicable to other diseases involving hypoxia or hypoxia with reoxygenation, such as sleep apnea, asthma, chronic obstructive pulmonary disease, cardiac arrest, and cardiopulmonary bypass.

Stroke models have also been used to study how drug delivery mechanisms may differ in the post-stroke BBB or how potential therapeutics affect BBB disruption in stroke [47-49]. Alterations in relevant BBB transporter systems, such as those for peptides and cytokines, or expression of other BBB proteins have also been studied in stroke or stroke-prone models [50-52]. Modeling can lead to therapeutic strategies such as increasing brain retention of potential therapeutics by inhibiting brain-to-blood efflux systems [53].

Stroke is usually considered a disease associated with aging, but one form, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), occurs in persons under 40 years of age [54]. CADASIL results from a mutation in the Notch3 gene. Although the mutation is throughout the vascular bed, it seems that only the central nervous system is affected. Mouse and pig models recapitulate various aspects of the disease, developing a CADASIL-like picture. Loss of pericytes is prerequisite to the development of altered BBB permeability and hypoperfusion (R Cecchelli, personal communication). Interestingly, pericyte drop out is also key to the develop of disruption of the blood-retinal barrier in diabetes mellitus [55;56].

Most models of diabetes mellitus use streptozotocin which destroys the beta cells of the pancreas. This produces an insulinopenic hyperglycemia and so models type I diabetes mellitus. However, as discussed below, aspects of the BBB are also affected in obesity. Most models of obesity are likely also producing animals with insulin resistance or hyperinsulinemic hyperglycemia. The latter would model diabetes mellitus type II. These models provide new opportunities to study the effects of type II diabetes mellitus on the BBB.

The BBB can be involved indirectly in models of diseases not typically thought to have a BBB component. For example, the lysosomal storage diseases result from defects in enzyme production, function, or translocation, allowing the accumulation of various storage products [57]. Dysfunction of both peripheral tissues and the CNS occurs with the accumulation of these storage products. In many of these diseases, normal function can be returned to the peripheral tissues by treatment with enzyme. However, these enzymes are large and do not cross the BBB of adults. As such, lysosomal storage diseases represent a major challenge for CNS drug delivery [58;59]. They also represent an excellent model for the development of drug delivery systems for large proteins. Animal models with the enzyme of interest knocked out will give a clear signal of clearing of storage product when therapeutic amounts of the enzyme are delivered to the brain. Most drug development for this disease has concentrated on delivering enzyme as a cargo attached to some other substance that will cross the BBB rapidly, the oft-called Trojan horse approach. More recent work has noted that the perinatal BBB expresses function mannose-6 phosphate receptor which transports into the brain two enzymes, GUSB whose deficiency causes Sly syndrome and sulfamidase whose deficiency causes Sanfilippo syndrome [60;61]. Thus, the brains of neonates in which these enzymes are knocked out can be successfully treated by giving peripheral enzyme [62-64]. However, the transporter function is lost with maturity and is virtually absent by adulthood. Recent work has shown that epinephrine can re-induce transporter function in adults [65]. Thus, lysosomal storage diseases may provide an excellent model for the study of transcytotic events and their induction at the BBB. Recent work has also suggested that the BBB is not itself resistant to the pathologies of lysosomal storage disease (DJ Begley, personal communication). Knockouts for MPS IIIA have a decreased brain vascular space, altered penetration of diazepam, and increased penetration of glycine. This opens the possibility that among the targets of these diseases is the BBB itself.

Functions of the BBB

Traditional studies have focused on BBB disruption. Classically, diseases such as multiple sclerosis, stroke/ischemia, trauma, Alzheimer’s disease, epilepsy, CNS cancers, hypertensive encephalopathy, and sepsis have been modeled [21;66-69]. More recently, neuroAIDs, diabetes mellitus, and chronic pain syndromes have been shown to have disruptions in the BBB [70-72].

A wide variety of approaches have been used to study BBB disruption and these have given different aspects to the overall picture. Injection of dyes was the original method used by Erlich in the 1880’s that led eventually to the elucidation of barrier function and was used for decades [73]. This method has enjoyed a resurgence in use in the last decade or so, possibly because little special equipment or regulatory oversight is needed. The method, however, requires skill and practice to give true results. Main sources of error are inadequate washout of the vascular space, disruption of the BBB from washout of the vascular space, or administration of too high a dose of dye. The latter occurs when the amount of dye given exceeds the binding capacity of serum albumin. These common errors all act to overestimate BBB disruption. Ultrastructural studies ultimately showed the anatomical basis for the BBB [74] and subsequently showed that most disruptions of the BBB [75] result from increased vesicular pathways (transcytosis) with or without an accompanying opening of tight junctions (paracellular pathways). Nevertheless, many current studies, especially those devoted to use of in vitro methods, focus on tight junctions. Radioactive studies using a series of molecular weight markers can often distinguish between disruption from transcytosis, in which the measure of disruption is similar regardless of the molecular size of the marker, vs paracellular mechanisms, in which smaller molecular weight markers often give larger measures of disruption [76;77].

In comparison to disruption, other aspects of BBB function during disease are relatively understudied. This is unfortunate, as it is likely that these other functions change much more early in the course of disease and likely are much more relevant to all but the terminal aspects of disease progression than is disruption. Changes in BBB lipid composition, receptor binding kinetics and function, transporter function, immune cell trafficking, enzymatic activity, and secretory capacity have been shown to change in models of aging, diabetes mellitus, sepsis, neuroimmune activation, multiple sclerosis, neuroAIDS, and Alzheimer’s disease [6;9;21;23;78-86]. Changes in transporter function as central to progression of obesity and Alzheimer’s disease is discussed below, but other important examples exist. For example, whereas disruption of the BBB begins after several weeks of streptozotocin-induced diabetes mellitus, the transport of insulin across the BBB is altered within 72 h [72;87]. Whereas the BBB can become disrupted in neuroAIDS or with seizures, these conditions cause alterations in p-glycoprotein expression [88;89]. This, in turn, alters the ability of anti-virals and anti-epileptic mediations to accumulate within the CNS.

Modeling of many diseases shows that the BBB is involved or altered on many levels [90]. For example, in neuroAIDS: the BBB is minimally disrupted [25;42;91]; activation of BBB cells and immune cells leads to enhanced adhesion of immune cells to the BBB and enhanced viral entry [25;33;34;92;93]; infected immune cells, free virus, and viral proteins including the neurotoxic gp120 and TAT cross the BBB [23;26;33;94;95]; BBB surface glycoprotein composition likely determines which HIV-1 strains are taken up or transported by the BBB [29]; BBB efflux systems impair the accumulation by the brain of many antivirals [38;39;96]; BBB secretions and cell surface expression of immune-active cytokines and chemokines is increased by virus or viral proteins [32;97;98]. These changes affect many aspects of AIDS and neuroAIDS including disease initiation and progression, neuroimmune activation, and drug delivery. Such multi-level changes in the BBB also occur in models for diabetes mellitus and sepsis/neuroimmune activation.

Another example of the expanded understanding of the sorts of roles the BBB may play in diseases is provided by epilepsy. Classically, the BBB has been noted to be disrupted in seizures with such disruption affected by parameters such as aging and gender [99;100]. More recently, drug delivery has become of great interest with the realization that an efflux system located at the BBB termed p-glycoprotein retards the accumulation by brain of most anti-seizure medications [101;102]. Indeed, the some 30% of epileptic patients who are resistant to anti-seizure medications may be so because of overexpression of p-glycoprotein [103]. Even more recently, it has been shown that seizure activity increases expression of p-glycoprotein at the BBB, thus complicating control of epilepsy [88;89]. Finally, immune cell-BBB interactions may be key to turning experimental seizure into epilepsy. Seizure activity induces selectin expression at the BBB which increases immune cell adhesion and perhaps CNS trafficking [8]. Blockade of immune cell-BBB adhesion inhibits both induction of acute seizures and chronic recurrent seizure activity.

Diseases Caused by the BBB: The Case for Alzheimer’s and Obesity

Diseases in which the BBB plays a central or primary role could be those in which either the BBB is the original site of pathophysiology or in which altered BBB function is key to disease progression. Multiple sclerosis and stroke including CADASIL have been considered above. To this list can be added De Vivo’s disease, which includes familial seizures and mental retardation resulting from underexpression of the GLUT-1 transporter at the BBB [104]. Encephalopathy occurs when the BBB is disrupted during hypertensive emergencies; although formerly thought to be entirely caused by physical disruption from high pressure, more recent investigations suggest that activity of the renin-angiotensin-aldosterone system in general and of angiotensin II in particular may be important. In a broader sense, epilepsy resistance to anti-seizure medications, which occurs in about 30% of cases, is caused by overexpression of p-glycoprotein at the BBB, and CNS infections and infestations arise because of their ability to interact with the BBB in a way to negotiate their permeation into the CNS. Arguments which place the BBB in a causal role in Alzheimer’s disease and obesity are illustrative of the general concepts of this review.

Alzheimer’s Disease

Several hypotheses have been advanced that would involve the BBB/cerebrovasculature (table I) in AD. These include BBB disruption, defective glucose transport, and release of neurotoxins from the BBB. One of the most recent hypotheses is the neurovascular hypothesis [3]. This states that deficient efflux of amyloid ß peptide (Aß) from the brain contributes to its accumulation and toxicity within the CNS. That Aß is removed from the CNS by a rapid saturable efflux system has been well documented by several laboratories [105-108]. In addition, Aß efflux is decreased in both transgenic mice and the SAMP8, a mouse strain with a natural mutation that results in an age-dependent increase in Aß and cognitive defects [105;109;110].

Table I.

Theories Relating Alzheimer’s Disease to the Blood-brain Barrier and the Cerebrovasculature

Leaky Blood-brain Barrier [163]
Tortuous Capillary Bed [164]
Defective Glucose Transporters [83]
Microvascularopathy [165]
Cerebrovascular Degeneration [166]
Brain Endothelium and the Release of Neurotoxins [82]
Permeation of Aluminum-glutamate Complexes [167]
Inhibition of Brain Endothelial Cell Proliferation [168]
Decreased Vitamin B12 Transport [169]
Vascular Cognitive Impairment [170]
Altered Insulin Transport into Brain [171]
Decreased Cerebral Blood Flow [172]
Aß Formation of Ionophores [173]
Neurovascular Hypothesis: Decreased Aß Efflux from Brain [4]

Efflux of Aß from brain has been suggested to be mediated by low density lipoprotein receptor-related protein-1 (LRP-1) and by p-glycoprotein [107;111-113]. Aß is a ligand for liver LRP-1 which likely plays an important role in its clearance from blood. Immunohistochemistry studies show that LRP-1 levels decrease in brains of AD patients [4]. However, other studies suggest that the relations among LRP-1, Aß, and p-glycoprotein are complex and may involve other ligands or transporters [106;114]. A model in which LRP-1 at the BBB was knocked down with antisense given by intracerebroventricular infusion showed that brain Aß levels increased about 40% and mice developed cognitive impairments [115]. These results are consistent with a major role in AD for the BBB efflux of Aß by an LRP-1 dependent system.

Other work has suggested that blood-to-brain transport of blood-borne Aß could contribute to brain levels of Aß with the receptor for advanced glycation end products (RAGE) acting as the primary transporter [116;117]. The contribution that circulating Aß makes to CNS levels of Aß is not established and so the role of blood-to-brain transport is less clear. Findings suggest, however, that just as Aß efflux is decreased in AD, blood-to-brain transport is increased.

Given the evidence that the BBB transporters are altered to favor increased influx and decreased efflux of Aß, the question arises as to what dictates these changes. Isoforms of Aß are differentially transported by LRP-1 and w ith aging in the SAMP8, efflux of Aß1–42 is more significantly lost than efflux of Aß1–40 [105;118]. This would favor retention of the more toxic form of Aß1–42 with aging. Dimerization and complexing with other LRP-1 ligands also retards Aß efflux [119]. Models of inflammation induced by lipopolysaccharide also recapitulate the proposed findings in AD, increasing influx and decreasing efflux of Aß. Interestingly, administration of the nonsteroidal antiinflammatory drug indomethacin prevents the lipopolysacchride-induced impairment of Aß efflux [120]. This is consistent with the proposed beneficial role of indomethacin in AD [121;122].

Obesity

Obesity is characterized by resistance to leptin, a 17 kDa protein secreted by fat tissue [123]. Leptin crosses the BBB by way of a saturable transport system where it interacts with its receptors located throughout the brain [124]. Most studied of the CNS sites is the arcuate nucleus. Here, leptin acts through its receptor to inhibit orexigenic peptides such as neuropeptide Y and orexin and to stimulate anorexigenic peptides such as melanocortin [125]. Leptin inhibits feeding through these mechanisms and also increases caloric expenditure. These two factors together result in a decrease in fat mass. As fat mass decreases, so does leptin levels in blood and the CNS; this, in turn, decreases anorexia and caloric expenditure. Thus, a negative feedback loop is formed between adipose tissue and feeding/caloric expenditure. Resistance to these actions of leptin or an absence of leptin results in obesity.

Leptin interactions with the BBB have been studied in several animal models, especially those of obesity. The earliest studies were those of van Heek and [126;127] in two models of obesity prone mice. These two studies showed that as obesity increased, these mice passed through a phase of resistance to peripherally administered leptin while remaining sensitive to centrally administered leptin. This is consistent with a block in the blood-to-brain transfer of leptin and suggested that failure of transport at the BBB preceded that of resistance at the arcuate nucleus. Pharmacokinetic studies in an obesity of maturity model, the Koletsy rat, and several other obesity models showed transport of leptin across the BBB to be decreased in proportion to the degree of obesity [128-131]. Studies with the brain perfusion model showed that leptin resistance at the BBB is complex, induced by both circulating factors, such as leptin inducing its own self-inhibition, and longer term factors inducing down regulation [128;132].

Investigation of transport of leptin across the BBB in various models has revealed several interesting characteristics. The transport rate is not static but influenced by obesity, ovarian factors, starvation, triglycerides, insulin, glucose, and epinephrine [133-136]. Effects of lipopolysaccharide, cholecystokinin, and alcohol administration have also been studied [137-139]. The nature of the BBB transporter protein for leptin is of great interest. Most work has indicated that at the vascular BBB, the short form of the leptin receptor likely acts as the leptin transporter. However, the Koletsky rat does not express functional receptors, yet is still able to transport leptin across the BBB by a saturable mechanism[131;140;141]. Therefore, there is likely a leptin transporter at the BBB other than the short form. Megalin has been proposed as the leptin transporter at the choroid plexus [142].

The role of the BBB in modifying leptin actions in the CNS likely affects many of leptin’s other effects. Leptin has effects on immune function, bone density, cognition, breathing, reproduction, and other functions, most mediated through CNS sites [143-150]. Leptin has receptors throughout the brain as well, including on non-neuronal cells such as astrocytes [151], so many of these actions may be mediated through different CNS locales and cell types. The rate of transport of leptin across the BBB varies among brain regions [152]. As such, the regional differences obesity has on transport likely means that obesity has differential effects on the leptin actions mediated through the CNS. Leptin also has functioning receptors within the subfornical organ, a circumventricular organ [153]. Similarly, there is evidence for some arcuate nucleus neurons projecting into the median eminence [154]. This suggests a dynamic interaction of CNS leptin receptor sites that are inside and outside the BBB as has been proposed for other peptides and regulatory proteins such as amylin and interleukin.

BBB interactions with other peptides and proteins related to feeding and metabolism have been studied in various models of disease. Starvation, immune activation with lipopolysacchride, obesity, and hypertriglyceridemic models have been used to study ghrlein or insulin transport [87;155-160]. Urocortin transport across the BBB is induced by leptin and tumor necrosis factor-alpha administration[161;162].

Conclusions

Our understanding of the BBB continues to evolve. As it does, the BBB is discovered to be involved ever more intimately in an ever wider array of diseases. This understanding has been greatly fostered and defined by models of the BBB and models of disease states. As these models continue to conceptualize the BBB not only as a physical barrier but also as a regulatory interface between the CNS and blood, more roles in disease states will likely be discovered. Most exciting are a series of diseases in which the BBB and its dysfunctions play key roles to disease onset and development.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Reference List

  • [1].Rapoport SI. Blood Brain Barrier in Physiology and Medicine. Raven Press; New York: 1976. [Google Scholar]
  • [2].Bradbury M. The Concept of a Blood-Brain Barrier. John Wiley and Sons Ltd; New York: 1979. [Google Scholar]
  • [3].Zlokovic BV. Neurovascular mechanisms of Alzheimer’s neurodegeneration. Trends Neurosci. 2005;28:202–208. doi: 10.1016/j.tins.2005.02.001. [DOI] [PubMed] [Google Scholar]
  • [4].Deane R, Sagare A, Zlokovic B. The role of the cell surface LRP and soluble LRP in blood-brain barrier Aß clearance in Alzheimer’s disease. Current Pharmaceutical Design. 2008;14:1601–1605. doi: 10.2174/138161208784705487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Huber JD. Diabetes, cognitive function, and the blood-brain barrier. Current Pharmaceutical Design. 2008;14:1594–1600. doi: 10.2174/138161208784705441. [DOI] [PubMed] [Google Scholar]
  • [6].Horani MH, Mooradian AD. The effect of diabetes on the blood brain barrier. Current Pharmaceutical Design. 2003;9:833–840. doi: 10.2174/1381612033455314. [DOI] [PubMed] [Google Scholar]
  • [7].Banks WA. The blood-brain barrier as a cause of obesity. Current Pharmaceutical Design. 2008;14:1606–1614. doi: 10.2174/138161208784705496. [DOI] [PubMed] [Google Scholar]
  • [8].Fabene PF, Navarro Mora G, Martinello M, Rossi B, Mergio F, Ottoboni L, Bach S, Angiari S, Benati D, Chakir A, Zanetti L, Schio F, Osculati A, Marzola P, Nicolato E, Homeister JW, Xia L, Lowe JB, McEver R, Osculati F, Sbarbati A, Butcher EC, Constantin G. A role for leukocyte-endothelial adhesion mechanisms in epilepsy. Nature Medicine. 2008;14:1377–1383. doi: 10.1038/nm.1878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Engelhardt B. The blood-central nervous system barriers actively control immune cell entry into the central nervous system. Current Pharmaceutical Design. 2008;14:1555–1565. doi: 10.2174/138161208784705432. [DOI] [PubMed] [Google Scholar]
  • [10].MCCandless EE, Budde M, Lees JR, Dorsey D, Lyng E, Klein RS. IL-1R signaling within the central nervous system regulates CXCL12 expression at the blood-brain barrier and disease severity during experimental autoimmune encephalomyelitis. J Immunology. 2009;183:613–620. doi: 10.4049/jimmunol.0802258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Schulze-Topphoff U, Prat A, Prozorovski T, Siffrin V, Paterka M, Herz J, Bendix I, Ifergan I, Schadock I, More MA, Van Horssen J, Schroter F, Smorodchenko A, Han MH, Bader M, Steinman L, Aktas O, Zipp F. Activiation of kinin receptor B1 limits encephalitogenic T lymphocyte recruitment to the central nervous system. Nat Med. 2009;15:788–793. doi: 10.1038/nm.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Baxter AG. The origin and application of experimental autoimmune encephalomyelitis. Nature Rev Immunol. 2007;7:904–912. doi: 10.1038/nri2190. [DOI] [PubMed] [Google Scholar]
  • [13].Lassmann H. Models of multiple sclerosis: new insights into pathophysiology and repair. Curr Opin Neurol. 2008;21:242–247. doi: 10.1097/WCO.0b013e3282fee94a. [DOI] [PubMed] [Google Scholar]
  • [14].McRae BL, Kennedy MK, Tan LJ, Dal Canto MC, Picha KS, Miller SD. Induction of active and adoptive relapsing experimental autoimmune encephalomyelitis (EAE) using an encephalitogenic epitope of proteolipid protein. J. Neuroimmunol. 1992;38:229–240. doi: 10.1016/0165-5728(92)90016-e. [DOI] [PubMed] [Google Scholar]
  • [15].Juhler M, Barry DI, Offner H, Konat G, Klinken L, Paulson OB. Blood-brain and blood-spinal cord barrier permeability during the course of experimental allergic encephalomyelitis in the rat. Brain Res. 1984;302:347–355. doi: 10.1016/0006-8993(84)90249-x. [DOI] [PubMed] [Google Scholar]
  • [16].Butter C, Baker D, O’Neill JK, Turk JL. Mononuclear cell trafficking and plasma protein extravasation into the CNS during chronic relapsing experimental allergic encephalomyelitis in Biozzi AB/H mice. J. Neurol. Sci. 1991;104:9–12. doi: 10.1016/0022-510x(91)90209-p. [DOI] [PubMed] [Google Scholar]
  • [17].Male D. The blood-brain barrier - No barrier to a determined lymphocyte. In: Greenwood J, Begley DJ, Segal MB, editors. New Concepts of a Blood-Brain Barrier. Plenum Press; New York: 1995. pp. 311–314. [Google Scholar]
  • [18].Lossinsky AS, Pluta R, Song MJ, Badmajew V, Moretz RC, Wisniewski HM. Mechanisms of inflammatory cell attachment in chronic relapsing experimental allergic encephalomyelitis: a scanning and high-voltage electron microscopic study of the injured mouse blood-brain barrier. Microvasc. Res. 1991;41:299–310. doi: 10.1016/0026-2862(91)90030-f. [DOI] [PubMed] [Google Scholar]
  • [19].Wolburg H, Wolburg-Buchholz K, Engelhardt B. Diapedesis of mononuclear cells across cerebral venules during experimental autoimmune encephalomyelitis leaves tight junctions intact. Acta Neuropathol. 2005;109:181–190. doi: 10.1007/s00401-004-0928-x. [DOI] [PubMed] [Google Scholar]
  • [20].Knobler RL, Marini JC, Goldowitz D, Lublin FD. Distribution of the blood-brain barrier in heterotopic brain transplants and its relationship to the lesions of EAE. J. Neuropathol. Exp. Neurol. 1992;51:36–39. doi: 10.1097/00005072-199201000-00005. [DOI] [PubMed] [Google Scholar]
  • [21].Pan W, Banks WA, Kennedy MK, Gutierrez EG, Kastin AJ. Differential permeability of the BBB in acute EAE: enhanced transport of TNF-α. Am. J. Physiol. 1996;271:E636–E642. doi: 10.1152/ajpendo.1996.271.4.E636. [DOI] [PubMed] [Google Scholar]
  • [22].Popovich PG, Stokes BT, Whitacre CC. Concept of autoimmunity following spinal cord injury: possible roles for T lymphocytes in the traumatized central nervous system. J. Neurosci. Res. 1996;45:349–363. doi: 10.1002/(SICI)1097-4547(19960815)45:4<349::AID-JNR4>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • [23].Persidsky Y, Stins M, Way D, Witte MH, Weinand M, Kim KS, Bock P, Gendelman HE, Fiala M. A model for monocyte migration through the blood-brain barrier during HIV-1 encephalitis. J. Immunol. 1997;158:3499–3510. [PubMed] [Google Scholar]
  • [24].Hurwitz AA, Berman JW, Lyman WD. The role of the blood-brain barrier in HIV infection of the central nervous system. Adv. Neuroimmunol. 1994;4:249–256. doi: 10.1016/s0960-5428(06)80263-9. [DOI] [PubMed] [Google Scholar]
  • [25].Toborek M, Lee YW, Flora G, Pu H, Andreeff M, Wylegala E, Henning B, Nath A. Mechanisms of the blood-brain barrier disruption in HIV-1 infection. Cell. Mol. Neurobiol. 2005;25:181–199. doi: 10.1007/s10571-004-1383-x. [DOI] [PubMed] [Google Scholar]
  • [26].Banks WA, Freed EO, Wolf KM, Robinson SM, Franko M, Kumar VB. Transport of human immunodeficiency virus type 1 pseudoviruses across the blood-brain barrier: role of envelope proteins and adsorptive endocytosis. J. Virol. 2001;75:4681–4691. doi: 10.1128/JVI.75.10.4681-4691.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Banks WA, Robinson SM, Wolf KM, Bess JW, Jr., Arthur LO. Binding, internalization, and membrane incorporation of human immunodeficiency virus-1 at the blood-brain barrier is differentially regulated. Neuroscience. 2004;128:143–153. doi: 10.1016/j.neuroscience.2004.06.021. [DOI] [PubMed] [Google Scholar]
  • [28].Banks WA, Akerstrom V, Kastin AJ. Adsorptive endocytosis mediates the passage of HIV-1 across the blood-brain barrier: evidence for a post-internalization coreceptor. J. Cell Sci. 1998;111:533–540. doi: 10.1242/jcs.111.4.533. [DOI] [PubMed] [Google Scholar]
  • [29].Bobardt MD, Salmon P, Wang L, Esko JD, Gabuzda D, Fiala M, Trono D, Van der Schueren B, David G, Gallay PA. Contribution of proteoglycans to human immunodeficiency virus type 1 brain invasion. J. Virol. 2004;78:6567–6584. doi: 10.1128/JVI.78.12.6567-6584.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Nakaoke R, Ryerse JS, Niwa M, Banks WA. Human immunodeficiency virus type 1 transport across the in vitro mouse brain endothelial cell monolayer. Exp. Neurol. 2005;193:101–109. doi: 10.1016/j.expneurol.2004.11.020. [DOI] [PubMed] [Google Scholar]
  • [31].Dohgu S, Banks WA. Lipopolysaccharide-enhanced transcellular transport of HIV-1 across the blood-brain barrier is medited by the p38 mitogen-activated protein kinase pathway. Exp. Neurol. 2008;210:740–749. doi: 10.1016/j.expneurol.2007.12.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Sanders VJ, Pittman CA, White MG, Wang G, Wiley CA, Achim CL. Chemokines and receptors in HIV encephalitis. AIDS. 1998;12:1021–1026. [PubMed] [Google Scholar]
  • [33].Nottet HS, Persidsky Y, Sasseville VG, Nukuna AN, Bock P, Zhai QH, Sharer LR, McComb RD, Swindells S, Soderland C, Gendelman HE. Mechanisms for the transendothelial migration of HIV-1-infected monocytes into brain. J. Immunol. 1996;156:1284–1295. [PubMed] [Google Scholar]
  • [34].Miller DW. Immunobiology of the blood-brain barrier. J. Neurovirology. 1999;5:570–578. doi: 10.3109/13550289909021286. [DOI] [PubMed] [Google Scholar]
  • [35].Ronaldson PT, Persidsky Y, Bendayan R. Regulation of ABC membrane transporters in glial cells: relevance to the pharmacotherapy of brain HIV-1 infection. GLIA. 2008;56:1711–1735. doi: 10.1002/glia.20725. [DOI] [PubMed] [Google Scholar]
  • [36].Zidovetzki R, Wang JL, Chen P, Jeyasseelan R, Hofman F. Human immunodeficiency virus Tat protein induces interleukin 6 mRNA expression in human brain endothelial cells via protein kinase C- and cAMP-dependent protein kinase pathways. AIDS Res. Hum. Retroviruses. 1998;14:825–833. doi: 10.1089/aid.1998.14.825. [DOI] [PubMed] [Google Scholar]
  • [37].Belliard AM, Tardivel S, Farinotti R, Lacour B, Leroy C. Effect of hr-IL-2 treatment on intestinal P-glycoprotein expression and activity in Caco-2 cells. J. Pharm. Pharmacol. 2002;54:1103–1109. doi: 10.1211/002235702320266262. [DOI] [PubMed] [Google Scholar]
  • [38].Lee CGL, Gottesman MM, Cardarelli CO, Ramachandra M, Jeang KT, Ambudkar SV, Pastan I, Dey S. HIV-1 protease inhibitors are substrates for the MDR1 multidrug transporter. Biochemistry. 1998;37:3594–3601. doi: 10.1021/bi972709x. [DOI] [PubMed] [Google Scholar]
  • [39].Thomas SA. Anti-HIV drug distribution to the central nervous system. Current Pharmaceutical Design. 2004;10:1313–1324. doi: 10.2174/1381612043384835. [DOI] [PubMed] [Google Scholar]
  • [40].Banks WA. Physiology and pathophysiology of the blood-brain barrier: Implications for microbial pathogenesis, drug delivery and neurodegenerative disorders. J. Neurovirology. 1999;5:538–555. doi: 10.3109/13550289909021284. [DOI] [PubMed] [Google Scholar]
  • [41].Yang WX, Terasaki T, Shiroki K, Ohka S, Aoki J, Tanabe S, Nomura T, Terada E, Sugiyama Y, Nomoto A. Efficient delivery of circulating poliovirus to the central nervous system independently of poliovirus receptor. Virology. 1997;229:421–428. doi: 10.1006/viro.1997.8450. [DOI] [PubMed] [Google Scholar]
  • [42].Chou S, Dix RD. Viral infections and the blood-brain barrier. In: Neuwelt EA, editor. Implications of the Blood-Brain Barrier and Its Manipulation, Volume 2: Clinical Aspects. Plenum Publishing Corporation; New York: 1989. pp. 449–468. [Google Scholar]
  • [43].Kim KS. Microbial translocation of the blood-brain barrier. Int J Parasitol. 2007;36:607–614. doi: 10.1016/j.ijpara.2006.01.013. [DOI] [PubMed] [Google Scholar]
  • [44].Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nat Med. 2004;10:1366–1373. doi: 10.1038/nm1140. [DOI] [PubMed] [Google Scholar]
  • [45].Brown RC, Davis TP. Calcium modulation of adherens tight junction function: a potential mechanism for blood-brain barrier disruption after stroke. Stroke. 2002;33:1706–1711. doi: 10.1161/01.str.0000016405.06729.83. [DOI] [PubMed] [Google Scholar]
  • [46].Pulsinelli WA, Brierly JB. A new model of bilateral hemispheric ischemia in the unanesthetized rat. Stroke. 1979;10:267–272. doi: 10.1161/01.str.10.3.267. [DOI] [PubMed] [Google Scholar]
  • [47].Chi OZ, Liu X, Weiss HR. Effects of 17beta-estradiol on blood-brain barrier disruption during focal ischemia in rats. Horm. Met. Res. 2003;34:530–534. doi: 10.1055/s-2002-34794. [DOI] [PubMed] [Google Scholar]
  • [48].Ehrenreich H, Hasselblatt M, Dembowski C, Depek L, Lewczuk P, Stiefel M, Rustenbeck H-H, Breiter N, Jacob S, Knerlich F, Bohn M, Poser W, Rither E, Kochen M, Gefeller O, Gleiter C, Wessel TC, De Ryck M, Itri L, Prange H, Cerami A, Brines M, Siren A-L. Erythropoietin therapy for acute stroke is both safe and beneficial. Molecular Medicine. 2002;8:495–505. [PMC free article] [PubMed] [Google Scholar]
  • [49].Pan Y, Zhang H, Acharya AB, Patrick PH, Oliver D, Morley JE. Effect of testosterone on functional recovery in a castrate male rat stroke model. Brain Res. 2005;1043:195–204. doi: 10.1016/j.brainres.2005.02.078. [DOI] [PubMed] [Google Scholar]
  • [50].Kirsch T, Wellner M, Luft FC, Haller H, Lippoldt A. Altered gene expression in cerebral capillaries of stroke-prone spontaneously hypertensive rats. Brain Res. 2001;910:106–115. doi: 10.1016/s0006-8993(01)02670-1. [DOI] [PubMed] [Google Scholar]
  • [51].Pan W, Ding Y, Yu Y, Ohtaki H, Nakamachi T, Kastin AJ. Stroke upregulates TNFalpha transport across the blood-brain barrier. Exp. Neurol. 2006;198:222–233. doi: 10.1016/j.expneurol.2005.11.020. [DOI] [PubMed] [Google Scholar]
  • [52].Somogyvari-Vigh A, Pan W, Reglodi D, Kastin AJ, Arimura A. Effect of middle cerebral artery occulsion on the passage of pituitary adenylate cyclase activating polypeptide across the blood-brain barrier in the rat. Regul. Pept. 2000;91:89–95. doi: 10.1016/s0167-0115(00)00123-3. [DOI] [PubMed] [Google Scholar]
  • [53].Dogrukol-Ak D, Kumar VB, Ryerse JS, Farr SA, Verma S, Nonaka K, Nakamachi T, Ohtaki H, Niehoff ML, Edwards JC, Shioda S, Morley JE, Banks WA. Isolation of peptide transport system-6 from brain endothelial cells: therapeutic effects with antisense inhibition in Alzheimer’s and stroke models. J Cereb Blood Flow Metab. 2009;29:411–422. doi: 10.1038/jcbfm.2008.131. [DOI] [PubMed] [Google Scholar]
  • [54].Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser MG. Cadasil. Lancet Neurology. 2009;8:643–653. doi: 10.1016/S1474-4422(09)70127-9. [DOI] [PubMed] [Google Scholar]
  • [55].Romeo G, Liu WH, Asnaghi V, Kern TS, Lorenzi M. Activation of nuclear factor-kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes. 2002;51:2241–2248. doi: 10.2337/diabetes.51.7.2241. [DOI] [PubMed] [Google Scholar]
  • [56].Addison DJ, Garner A, Ashton N. Degeneration of intramural pericytes in diabetic retinopathy. Br. Med. J. 1970;1:264–266. doi: 10.1136/bmj.1.5691.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Begley DJ, Pontikis CC, Scarpa M. Lysosomal storage diseases and the blood-brain barrier. Current Pharmaceutical Design. 2008;14:1566–1580. doi: 10.2174/138161208784705504. [DOI] [PubMed] [Google Scholar]
  • [58].Boado RJ. A new generation of neurobiological drugs engineered to overcome the challenges of brain drug delivery. Drug News Perspective. 2008;21:489–503. doi: 10.1358/dnp.2008.21.9.1290820. [DOI] [PubMed] [Google Scholar]
  • [59].De Boer AG, Gaillard PJ. Drug targeting to the brain. Annu Rev Pharmacol Toxicol. 2007;47:323–355. doi: 10.1146/annurev.pharmtox.47.120505.105237. [DOI] [PubMed] [Google Scholar]
  • [60].Urayama A, Grubb JH, Sly WS, Banks WA. Developmentally regulated mannose 6-phosphate receptor-mediated transport of a lysosomal enzyme across the blood-brain barrier. Proc. Natl. Acad. Sci. USA. 2004;101:12658–12663. doi: 10.1073/pnas.0405042101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Urayama A, Grubb JH, Sly WS, Banks WA. Mannose 6-phosphate receptor mediated transport of sulfamidase across the blood-brain barrier in the newborn mouse. Molecular Therapy. 2008;16:1261–1266. doi: 10.1038/mt.2008.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Vogler C, Levy B, Galvin NJ, Thorpe C, Sands MS, Barker JE, Baty J, Birkenmeier EH, Sly WS. Enzyme replacement in murine mucopolysaccharidosis type VII: neuronal and glial response to beta-glucuronidase requires early initiation of enzyme replacement therapy. Pediatric Research. 1999;45:838–844. doi: 10.1203/00006450-199906000-00010. [DOI] [PubMed] [Google Scholar]
  • [63].Auclair D, Hopwood JJ, Brooks DA, Lemontt JF, Crawley AC. Replacement therapy for mucopolysaccharidosis type VI: advantages of early onset of therapy. Mol. Genet. Metab. 2003;78:163–174. doi: 10.1016/s1096-7192(03)00007-6. [DOI] [PubMed] [Google Scholar]
  • [64].Gliddon BL, Hopwood JJ. Enzyme-replacement therapy from birth delays the development of behavior and learning problems in mucopolysaccharidosis Type IIIA mice. Pediatric Research. 2004;56:1–8. doi: 10.1203/01.PDR.0000129661.40499.12. [DOI] [PubMed] [Google Scholar]
  • [65].Urayama A, Grubb JH, Banks WA, Sly WS. Epinephrine enhances lysosomal enzyme delivery across the blood-brain barrier by up-regulation of the mannos 6-phosphate receptor. Proc. Natl. Acad. Sci. USA. 2007;31:12873–12878. doi: 10.1073/pnas.0705611104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Abraham CS, Harada N, Deli MA, Niwa M. Transient forebrain ischemia increases the blood-brain barrier permeability for albumin in stroke-prone spontaneously hypertensive rats. Cell. Mol. Neurobiol. 2003;22:455–462. doi: 10.1023/a:1021067822435. [DOI] [PubMed] [Google Scholar]
  • [67].Baldwin SA, Fugaccia I, Brown DR, Brown LV, Scheff SW. Blood-brain barrier breach following cortical contusion in the rat. J. Neurosurg. 1996;85:476–481. doi: 10.3171/jns.1996.85.3.0476. [DOI] [PubMed] [Google Scholar]
  • [68].Pan W, Kastin AJ, Pick CG. The staircase test in mice after spinal cord injury. International Journal of Neuroprotection and Neuroregeneration. 2005;1:32–37. [Google Scholar]
  • [69].Pozzilli C, Bernardi S, Mansi L, Picozzi P, Iannotti F, Alfano B, Bozzao L, Lenzi GL, Salvatore M, Conforti P, Fieschi C. Quantitative assessment of the blood-brain barrier permeability in multiple sclerosis using 68-Ga-EDTA and positron emission tomography. J. Neurol. Neurosurg. Psych. 1988;51:1058–1062. doi: 10.1136/jnnp.51.8.1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Boven LA, Middel J, Verhoef J, De Groot CJ, Nottet HS. Monocytes infiltration is highly associated with loss of tight junction protein zonula occludens in HIV-1-associated dementia. Neuropathology and Applied Neurobiology. 2000;26:356–362. doi: 10.1046/j.1365-2990.2000.00255.x. [DOI] [PubMed] [Google Scholar]
  • [71].Brooks TA, Hawkins BT, Huber JD, Egleton RD, Davis TP. Chronic inflammatory pain leads to increased blood-brain barrier permeability and tight junction protein alterations. Am J Physiology. 2005;289:H738–H743. doi: 10.1152/ajpheart.01288.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Huber JD, VanGilder RL, Houser KA. Streptozotocin-induced diabetes progressively increases blood-brain barrier permeability in specific brain regions in rats. Am. J. Physiol. 2006;291:H2660–H2668. doi: 10.1152/ajpheart.00489.2006. [DOI] [PubMed] [Google Scholar]
  • [73].Broman T. Supravital analysis of disorders in the cerebrovascular permeability. Acta Psychiatr. 1950;25:19–31. doi: 10.1111/j.1600-0447.1950.tb04601.x. [DOI] [PubMed] [Google Scholar]
  • [74].Reese TS, Karnovsky MJ. Fine structural localization of a blood-brain barrier to exogenous peroxidase. J. Cell Biol. 1967;34:207–217. doi: 10.1083/jcb.34.1.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Lossinsky AS, Vorbrodt AW, Wisniewski HM. Ultracytochemical studies of vesicular and canalicular transport structures in the injured mammalian blood-brain barrier. Acta Neuropathol. (Berl) 1983;61:239–245. doi: 10.1007/BF00691992. [DOI] [PubMed] [Google Scholar]
  • [76].Ziylan YZ, Robinson PJ, Rapoport SI. Blood-brain barrier permeability to sucrose and dextran after osmotic opening. Am. J. Physiol. 1984;247:R634–R638. doi: 10.1152/ajpregu.1984.247.4.R634. [DOI] [PubMed] [Google Scholar]
  • [77].Mayhan WG, Heistad DD. Permeability of blood-brain barrier to various sized molecules. Am J Physiology. 1985;248:H712–H718. doi: 10.1152/ajpheart.1985.248.5.H712. [DOI] [PubMed] [Google Scholar]
  • [78].Banks WA, Farr SA, Morley JE. Permeability of the blood-brain barrier to albumin and insulin in the young and aged SAMP8 mouse. Journal of Gerontology: Biological Science. 2000;55A:B601–B606. doi: 10.1093/gerona/55.12.b601. [DOI] [PubMed] [Google Scholar]
  • [79].Daniel PM, Love ER, Pratt OE. The effect of age upon the influx of glucose into the brain. J. Physiol. (London) 1978;274:141–148. doi: 10.1113/jphysiol.1978.sp012139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].McLay RN, Kastin AJ, Zadina JE. Passage of interleukin-1-beta across the blood-brain barrier is reduced in aged mice: A possible mechanism for diminished fever in aging. nim. 2000;8:148–153. doi: 10.1159/000054275. [DOI] [PubMed] [Google Scholar]
  • [81].Mooradian AD, Morin AM, Cipp LJ, Haspel HC. Glucose transport is reduced in the blood-brain barrier of aged rats. Brain Res. 1991;551:145–149. doi: 10.1016/0006-8993(91)90926-m. [DOI] [PubMed] [Google Scholar]
  • [82].Grammas P, Botchlet TR, Moore P, Weigel PH. In: Production of neurotoxic factors by brain endothelium in Alzheimer’s disease. de la Torre JC, Hachinski V, editors. Annals of the New York Academy of Sciences; New York, NY: 1997. pp. 47–55. 826. Ref Type: Serial (Book,Monograph)
  • [83].Harik SI, Kalaria RN. Blood-brain barrier abnormalities in Alzheimer’s disease. Ann. NY Acad. Sci. 1991;640:47–52. doi: 10.1111/j.1749-6632.1991.tb00189.x. [DOI] [PubMed] [Google Scholar]
  • [84].Banks WA, Dohgu S, Nakaoke R, Lynch JL, Fleegal-DeMotta MA, Erickson MA, Vo TQ. Nitric oxide isoenzymes regulate LPS-enhanced insulin transport across the blood-brain barrier. Endocrinology. 2008;149:1514–1523. doi: 10.1210/en.2007-1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Reyes TM, Fabry Z, Coe CL. Brain endothelial cell production of a neuroprotective cytokine, interleukin-6, in response to noxious stimuli. Brain Res. 1999;851:215–220. doi: 10.1016/s0006-8993(99)02189-7. [DOI] [PubMed] [Google Scholar]
  • [86].Salkeni MA, Lynch JL, Price TO, Banks WA. Lipopolysaccharide impairs blood-brain barrier P-glycoprotein function in mice through prostaglandin- and nitric oxide-independent pathways and nitric oxide-independent pathways. J Neuroimmune Pharmacology. 2009;4:276–282. doi: 10.1007/s11481-008-9138-y. J Neuroimmune Pharmacology. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Banks WA, Jaspan JB, Kastin AJ. Effect of diabetes mellitus on the permeability of the blood-brain barrier to insulin. Peptides. 1997;18:1577–1584. doi: 10.1016/s0196-9781(97)00238-6. [DOI] [PubMed] [Google Scholar]
  • [88].Rizzi M, Caccia S, Guiso G, Richichi C, Gorter JA, Aronica E, Aliprandi M, Bagnati R, Fanelli R, D’Incalci M, Samanin R, Vezzani A. Limbic seizures induce P-glycoprotein in rodent brain: functional implications for pharmacoresistance. J. Neurosci. 2002;22:5833–5839. doi: 10.1523/JNEUROSCI.22-14-05833.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Lazarowski A, Ramos AJ, Garcia-Rivello H, Brusco A, Girardi E. Neuronal and glial expression of the multidrug resistance gene product in an experimental epilepsy model. Cell. Mol. Neurobiol. 2004;24:77–85. doi: 10.1023/b:cemn.0000012726.43842.d2. [DOI] [PubMed] [Google Scholar]
  • [90].Banks WA. Protector, prey, or perpetrator: The pathophysiology of the blood-brain barrier in NeuroAIDS. Science Online: NeuroAIDS. 2000;3:1–6. [Google Scholar]
  • [91].Avison MJ, Nath A, Greene-Avison R, Schmitt FA, Bales RA, Ethisham A, Greenberg RN, Berger JR. Inflammatory changes and breakdown of microvascular integrity in early human immunodeficiency virus dementia. Journal of Neurovirology. 2004;10:223–232. doi: 10.1080/13550280490463532. [DOI] [PubMed] [Google Scholar]
  • [92].Fiala M, Looney DJ, Stins M, Way DD, Zhang L, Gan X, Chiappelli F, Schweitzer ES, Shapshak P, Weinand M, Graves MC, Witte M, Kim KS. TNF-α opens a paracellular route for HIV-1 invasion across the blood-brain barrier. Molecular Medicine. 1997;3:553–564. [PMC free article] [PubMed] [Google Scholar]
  • [93].Stins MF, Shen Y, Huang SH, Gilles F, Kalra VK, Kim KS. Gp120 activates children’s brain endothelial cells via CD4. J Neurovirol. 2001;7:125–134. doi: 10.1080/13550280152058780. [DOI] [PubMed] [Google Scholar]
  • [94].Banks WA, Kastin AJ, Akerstrom V. HIV-1 protein gp120 crosses the blood-brain barrier: role of adsorptive endocytosis. Life Sci. 1997;61:L119–L125. doi: 10.1016/s0024-3205(97)00597-3. [DOI] [PubMed] [Google Scholar]
  • [95].Banks WA, Robinson SM, Nath A. Permeability of the blood-brain barrier to HIV-1 Tat. Exp. Neurol. 2005;193:218–227. doi: 10.1016/j.expneurol.2004.11.019. [DOI] [PubMed] [Google Scholar]
  • [96].Masereeuw R, Jaehde U, Langemeijer MWE, De Boer AG, Breimer DD. In vivo and in vitro transport of zidovudine (AZT) across the blood-brain barrier and the effects of transport inhibitors. Pharm. Res. 1994;11:324–330. doi: 10.1023/a:1018932213953. [DOI] [PubMed] [Google Scholar]
  • [97].Zidovetzki R, Wang JL, Chen P, Jeyasseelan R, Hofman F. Human immunodeficiency virus Tat protein induces interleukin 6 mRNA expression in human brain endothelial cells via protein kinase C- and cAMP-dependent protein kinase pathways. AIDS Res. Hum. Retroviruses. 1998;14:825–833. doi: 10.1089/aid.1998.14.825. [DOI] [PubMed] [Google Scholar]
  • [98].Shaw KT, Greig NH. Chemokine receptor mRNA expression at the in vitro blood-brain barrier during HIV infection. Neuroreport. 1999;10:53–56. doi: 10.1097/00001756-199901180-00010. [DOI] [PubMed] [Google Scholar]
  • [99].Oztas B, Kaya M, Camurcu S. Age related changes in the effect of electroconvulsive shock on the blood brain barrier permeability in rats. Mechanisms of Ageing and Development. 1990;51:149–155. doi: 10.1016/0047-6374(90)90097-y. [DOI] [PubMed] [Google Scholar]
  • [100].Oztas B, Kucuk M, Kaya M. Sex-dependent changes in blood-brain barrier permeability in epileptic rats following acute hyperosmotic exposure. Pharmacological Research. 2001;43:469–472. doi: 10.1006/phrs.2001.0797. [DOI] [PubMed] [Google Scholar]
  • [101].Begley DJ. ABC transporters and the blood-brain barrier. Current Pharmaceutical Design. 2004;10:1295–1312. doi: 10.2174/1381612043384844. [DOI] [PubMed] [Google Scholar]
  • [102].Tsuji A, Tamai I. Blood-brain barrier function of P-glycoprotein. Advanced Drug Delivery Reviews. 1997;25:287–298. [Google Scholar]
  • [103].Löscher W, Potschka H. Role of multidrug transporters in pharmacoresistance to antiepileptic drugs. J. Pharmacol. Exp. Ther. 2002;30:7–14. doi: 10.1124/jpet.301.1.7. [DOI] [PubMed] [Google Scholar]
  • [104].De Vivo DC, Trifiletti RR, Jacobson RI, Ronen GM, Behmand RA, Harik SI. Defective glucose transport across the blood-brain barrier as a cause of persistent hypoglycorrhachia, seizures, and developmental delay. N. Engl. J. Med. 1991;325:703–709. doi: 10.1056/NEJM199109053251006. [DOI] [PubMed] [Google Scholar]
  • [105].Banks WA, Robinson SM, Verma S, Morley JE. Efflux of human and mouse amyloid ß proteins 1–40 and 1–42 from brain: Impairment in a mouse model of Alzheimer’s disease. Neuroscience. 2003;121:487–492. doi: 10.1016/s0306-4522(03)00474-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Ito S, Ohtsuki S, Terasaki T. Functional characterization of the brain-to-blood efflux clearance of human amyloid-beta peptide (1–40) across the rat blood-brain barrier. Neurosci Res. 2006;56:246–252. doi: 10.1016/j.neures.2006.07.006. [DOI] [PubMed] [Google Scholar]
  • [107].Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, Holtzman DM, Miller CA, Strickland DK, Ghiso J, Zlokovic BV. Clearance of Alzheimer’s amyloid-β1–40 peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J. Clin. Invest. 2000;106:1489–1499. doi: 10.1172/JCI10498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Ghersi-Egea JF, Gorevic PD, Ghiso J, Frangione B, Patlak CS, Fenstermacher JD. Fate of cerebrospinal fluid-borne amyloid β-peptide: rapid clearance into blood and appreciable accumulation by cerebral arteries. J. Neurochem. 1996;67:880–883. doi: 10.1046/j.1471-4159.1996.67020880.x. [DOI] [PubMed] [Google Scholar]
  • [109].Kandimalla KK, Curran GL, Holasek SS, Gilles EJ, Wengenack TM, Poduslo JF. Pharmacokinetic analysis of the blood-brain barrier transport of 125I-amyloid ß protein 40 in wild-type and Alzheimer’s disease transgenic mic (APP,PSI) and its implications for amyloid plaque formation. J. Pharmacol. Exp. Ther. 2006;313:1370–1378. doi: 10.1124/jpet.104.081901. [DOI] [PubMed] [Google Scholar]
  • [110].LaRue B, Hogg E, Sagare A, Jovanovic S, Maness L, Maurer C, Deane R, Zlokovic B. Method for measurement of the blood-brain barrier permeability in the perfused mouse brain: application to amyloid-beta peptide in wild type and Alzheimer’s Tg2576 mice. J. Neurosci. Methods. 2004;138:233–244. doi: 10.1016/j.jneumeth.2004.04.026. [DOI] [PubMed] [Google Scholar]
  • [111].Vogelgesang S, Cascorbi I, Schroeder E, Pahnke J, Kroemer HK, Siegmund W, Kunert-Keil C, Walker LC, Warzok RW. Deposition of Alzheimer’s beta-amyloid is inversely correlated with p-glycoprotein expression in the brains of elderly non-demented humans. Pharmacogenetics. 2002;12:535–541. doi: 10.1097/00008571-200210000-00005. [DOI] [PubMed] [Google Scholar]
  • [112].Lam FC, Liu R, Shapiro AB, Renoir JM, Sharon FJ, Reiner PB. beta-Amyloid efflux mediated by p-glycoprotein. Journal of Neurochemisrty. 2001;76:1121–1128. doi: 10.1046/j.1471-4159.2001.00113.x. [DOI] [PubMed] [Google Scholar]
  • [113].Cirrito JR, Deane R, Fagan AM, Spinner ML, Parasadanian M, Finn MB, Jiang H, Prior JL, Sagare A, Bales KR, Paul SM, Zlokovic B, Piwnica-Worms D, Holztman DM. P-glycoprotein deficiency at the blood-brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model. J. Clin. Invest. 2005;115:3285–3290. doi: 10.1172/JCI25247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Yamada K, Hashimoto T, Yabuki C, Nagae Y, Tachikawa M, Strickland DK, Liu Q, Bu G, Basak JM, Holtzman DM, Ohtsuki S, Terasaki T, Iwatsubo T. The low density lipoprotein receptor-related protein 1 mediates uptake of amyloid beta peptides in an in vitro model of the blood-brain barrier cells. J Biol Chem. 2008;283:34554–34562. doi: 10.1074/jbc.M801487200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Jaeger JB, Dohgu S, Hwang MC, Farr SA, Murphy MP, Fleegal-DeMotta MA, Lynch JL, Robinson SM, Niehoff ML, Johnson SN, Kumar VB, Banks WA. Testing the neurovascular hypothesis of Alzheimer’s disease: LRP-1 antisense reduced blood-brain barrier clearance, increases brain levels of amyloid beta protein, and impairs cognition. J Alz Dis. 2009;17:553–570. doi: 10.3233/JAD-2009-1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].LaRue B, Hogg E, Sagare A, Jovanovic S, Maness L, Maurer C, Deane R, Zlokovic BV. Method for measurement of the blood-brain barrier permeability in the perfused mouse brain: application to amyloid-beta peptide in wild type and Alzheimer’s Tg2576 mice. Journal of Neuroscience Methods. 2004;138:233–242. doi: 10.1016/j.jneumeth.2004.04.026. [DOI] [PubMed] [Google Scholar]
  • [117].Deane R, Yan SD, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B. RAGE mediates amyloid-ß peptide transport across the blood-brain barrier and accumulation in brain. Nature Medicine. 2003;7:907–913. doi: 10.1038/nm890. [DOI] [PubMed] [Google Scholar]
  • [118].Deane R, Wu Z, Sagare A, Davis J, Du Yan S, Hamm K, Xu F, Parisi M, LaRue B, Hu HW, Spijkers P, Guo H, Song X, Lenting PJ, Van Nostrand WE, Zlokovic B. LRP/amyloid beta-peptide interaction mediates differential brain efflux of Abeta isoforms. Neuron. 2004;43:333–344. doi: 10.1016/j.neuron.2004.07.017. [DOI] [PubMed] [Google Scholar]
  • [119].Ito S, Ohtsuki S, Kamiie J, Nezu Y, Terasaki T. Cerebral clearance of human amyloid-beta peptide (1–40) across the blood-brain barrier is reduced by self-aggregation and formation of low-density lipoprotein receptor-related protein-1 ligand complexes. J Neurochem. 2007;103:2482–2490. doi: 10.1111/j.1471-4159.2007.04938.x. [DOI] [PubMed] [Google Scholar]
  • [120].Jaeger JB, Dohgu S, Lynch JL, Fleegal-DeMotta MA, Banks WA. Effects of lipopolysaccharide on the blood-brain barrier transport of amyloid beta protein: A mechanism for inflammation in the progression of Alzheimer’s disease. Brain,Behavior, and Immunity. 2009;23:507–517. doi: 10.1016/j.bbi.2009.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Szekely CA, Thorne JE, Zandi PP, Ek M, Messias E, Breitner JC, Goodman SN. Nonsteroidal anti-inflammatory drugs for the prevention of Alzheimer’s disease: A systematic review. Neuroepidemiology. 2004;23:159–169. doi: 10.1159/000078501. [DOI] [PubMed] [Google Scholar]
  • [122].Townsend KP, Pratico D. Novel therapeutic opportunities for Alzheimer’s disease: focus on nonsteroidal anti-inflammatory drugs. FASEB J. 2005;19:1592–1601. doi: 10.1096/fj.04-3620rev. [DOI] [PubMed] [Google Scholar]
  • [123].Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395:763–770. doi: 10.1038/27376. [DOI] [PubMed] [Google Scholar]
  • [124].Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM. Leptin enters the brain by a saturable system independent of insulin. Peptides. 1996;17:305–311. doi: 10.1016/0196-9781(96)00025-3. [DOI] [PubMed] [Google Scholar]
  • [125].Schwartz MW, Woods SC, Porte D, Jr., Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404:661–671. doi: 10.1038/35007534. [DOI] [PubMed] [Google Scholar]
  • [126].van Heek M, Compton DS, France CF, Tedesco RP, Fawzi AB, Graziano MP, Sybertz EJ, Strader CD, Davis J. Diet-induced obese mice develop peripheral, but not central, resistance to leptin. J. Clin. Invest. 1997;99:385–390. doi: 10.1172/JCI119171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Halaas JL, Boozer c., Blair-West J, Fidahusein N, Denton DA, Friedman JM. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc. Natl. Acad. Sci. USA. 1997;94:8878–8883. doi: 10.1073/pnas.94.16.8878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Banks WA, DiPalma CR, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity. Peptides. 1999;20:1341–1345. doi: 10.1016/s0196-9781(99)00139-4. [DOI] [PubMed] [Google Scholar]
  • [129].Burguera B, Couce ME, Curran GL, Jensen MD, Lloyd RV, Cleary MP, Poduslo JF. Obesity is associated with a decreased leptin transport across the blood-brain barrier in rats. Diabetes. 2000;49:1219–1223. doi: 10.2337/diabetes.49.7.1219. [DOI] [PubMed] [Google Scholar]
  • [130].Levin BE, Dunn-Meynell AA, Banks WA. Obesity-prone rats have normal blood-brain barrier transport but defective central leptin signaling before obesity onset. Am. J. Physiol. 2004;286:R143–R150. doi: 10.1152/ajpregu.00393.2003. [DOI] [PubMed] [Google Scholar]
  • [131].Kastin AJ, Pan W, Maness LM, Koletsky RJ, Ernsberger P. Decreased transport of leptin across the blood-brain barrier in rats lacking the short form of the leptin receptor. Peptides. 1999;20:1449–1453. doi: 10.1016/s0196-9781(99)00156-4. [DOI] [PubMed] [Google Scholar]
  • [132].Banks WA, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity is acquired and reversible. Am J Physiology. 2003;285:E10–E15. doi: 10.1152/ajpendo.00468.2002. [DOI] [PubMed] [Google Scholar]
  • [133].Banks WA, Coon AB, Robinson SM, Moinuddin A, Shultz JM, Nakaoke R, Morley JE. Triglycerides induce leptin resistance at the blood-brain barrier. Diabetes. 2004;53:1253–1260. doi: 10.2337/diabetes.53.5.1253. [DOI] [PubMed] [Google Scholar]
  • [134].Kastin AJ, Akerstrom V, Maness LM. Chronic loss of ovarian function decreases transport of leptin into mouse brain. Neurosci. Lett. 2001;310:69–71. doi: 10.1016/s0304-3940(01)02074-2. [DOI] [PubMed] [Google Scholar]
  • [135].Kastin AJ, Akerstrom V. Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice. Neuroendocrinology. 2001;73:237–242. doi: 10.1159/000054640. [DOI] [PubMed] [Google Scholar]
  • [136].Banks WA. Enhanced leptin transport across the blood-brain barrier by α1-adrenergic agents. Brain Res. 2001;899:209–217. doi: 10.1016/s0006-8993(01)02242-9. [DOI] [PubMed] [Google Scholar]
  • [137].Nonaka N, Hileman SM, Shioda S, Vo P, Banks WA. Effects of lipopolysaccharide on leptin transport across the blood-brain barrier. Brain Res. 2004;1016:58–65. doi: 10.1016/j.brainres.2004.04.066. [DOI] [PubMed] [Google Scholar]
  • [138].Pan W, Barron M, Hsuchou H, Tu H, Kastin AJ. Increased leptin permeation across the blood-brain barrier after chronic alcohol ingestion. Neuropsychopharmacology. 2008;33:859–866. doi: 10.1038/sj.npp.1301452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Cano V, Merino B, Ezquerra L, Somoza B, Ruiz-Gayo M. A cholecystokinin-1 receptor agonist (CCK-8) mediates increased permeability of brain barriers to leptin. Br J Pharmacol. 2008;154:1009–1015. doi: 10.1038/bjp.2008.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Banks WA, Niehoff ML, Martin D, Farrell CL. Leptin transport across the blood-brain barrier of the Koletsky rat is not mediated by a product of the leptin receptor gene. Brain Research. 2002;950:130–136. doi: 10.1016/s0006-8993(02)03013-5. Ref Type: Journal (Full)
  • [141].Wu-Peng XS, Chua SC, Jr., Okada N, Liu SM, Nicolson M, Leibel RL. Phenotype of the obese Koletsky (f) rat due to Tyr796Stop mutation in the extracellular domain of the leptin receptor (Lepr): evidence for deficient plasma-to-CSF transport of leptin in both the Zucker and Koletsky obese rat. Diabetes. 1997;46:513–518. doi: 10.2337/diab.46.3.513. [DOI] [PubMed] [Google Scholar]
  • [142].Dietrich MO, Spuch C, Antequerra D, Rodal I, deYebenes JG, Molina JA, Bermejo F, Carro E. Megalin mediates the transport of leptin across the blood-CSF barrier. Neurobiol Aging. 2008;29:902–912. doi: 10.1016/j.neurobiolaging.2007.01.008. [DOI] [PubMed] [Google Scholar]
  • [143].Ahima RS, Bjorbaek C, Osei S, Flier JS. Regulation of neuronal and glial proteins by leptin: implications for brain development. Endocrinology. 1999;140:2755–2762. doi: 10.1210/endo.140.6.6774. [DOI] [PubMed] [Google Scholar]
  • [144].Ahima RS, Dushay J, Flier SN, Prabakaran D, Flier JS. Leptin accelerates the onset of puberty in normal female mice. J. Clin. Invest. 1997;99:391–395. doi: 10.1172/JCI119172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Bell FT, Shen J, Vinson C, Rueger JM, Karsenty G. Leptin inhibits bone formation through a hypothalamic relay: A central control of bone mass. Cell. 2000;100:197–207. doi: 10.1016/s0092-8674(00)81558-5. [DOI] [PubMed] [Google Scholar]
  • [146].Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yand X, Liu X, Kondo H, Richards WG, Bannon TW, Noda M, Clement K, Vaisse C, Karsenty G. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature. 2005;434:514–520. doi: 10.1038/nature03398. [DOI] [PubMed] [Google Scholar]
  • [147].Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI. Leptin modulates the T-cell immune respone and reverses starvation-induced immunosuppression. Nature. 1999;394:897–901. doi: 10.1038/29795. [DOI] [PubMed] [Google Scholar]
  • [148].O’Donnell CP, Schaub CD, Haines AS, Berkowitz DE, Tankersley CG, Schwartz AR, Smith PL. Leptin prevents respiratory depression in obesity. Am. J. Respir. Crit. Care Med. 1999;159:1477–1484. doi: 10.1164/ajrccm.159.5.9809025. [DOI] [PubMed] [Google Scholar]
  • [149].Saaresranta T, Polo O. Does leptin link sleep loss and breathing disturbances with major public diseases? Ann. Med. 2004;36:172–183. doi: 10.1080/07853890310024659. [DOI] [PubMed] [Google Scholar]
  • [150].Farr SA, Banks WA, Morley JE. Effects of leptin on memory processing. Peptides. 2006;27:1420–1425. doi: 10.1016/j.peptides.2005.10.006. [DOI] [PubMed] [Google Scholar]
  • [151].Pan W, Hsuchou H, He Y, Sakharkar A, Cain C, Yu C, Kastin AJ. Astrocyte leptin receptor (ObR) and leptin transport in adult-onset obese mice. Endocrinology. 2008;149:2798–2806. doi: 10.1210/en.2007-1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Banks WA, Clever CM, Farrell CL. Partial saturation and regional variation in the blood to brain transport of leptin in normal weight mice. Am. J. Physiol. 2000;278:E1158–E1165. doi: 10.1152/ajpendo.2000.278.6.E1158. [DOI] [PubMed] [Google Scholar]
  • [153].Smith PM, Chambers AP, Price CJ, Ho W, Hopf C, Sharkey KA, Ferguson AV. The subfornical organ: a central nervous system site for actions of circulating leptin. Am J Physiol Regul Integr Comp Physiol. 2009;296:R512–R520. doi: 10.1152/ajpregu.90858.2008. [DOI] [PubMed] [Google Scholar]
  • [154].Faouzi M, Leshan R, Bjornholm M, Hennessey T, Jones J, Munzberg H. Differential accessibility of circulating leptin to individual hypothalamic sites. Endocrinology. 2007;148:5414–5423. doi: 10.1210/en.2007-0655. [DOI] [PubMed] [Google Scholar]
  • [155].Banks WA, Burney BO, Robinson SM. Effects of triglycerides, obesity, and starvation on ghrelin transport across the blood-brain barrier. Peptides. 2008;29:2061–2065. doi: 10.1016/j.peptides.2008.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Urayama A, Banks WA. Starvation and triglycerides reverse the obesity-induced impairment of insulin transport at the blood-brain barrier. Endocrinology. 2008;149:3592–3597. doi: 10.1210/en.2008-0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Xaio H, Banks WA, Niehoff ML, Morley JE. Effect of LPS on the permeability of the blood-brain barrier to insulin. Brain Res. 2001;896:36–42. doi: 10.1016/s0006-8993(00)03247-9. [DOI] [PubMed] [Google Scholar]
  • [158].Baskin DG, Stein LJ, Ikeda H, Woods SC, Figlewicz DP, Porte D, Jr., Greenwood MR, Dorsa DM. Genetically obese Zucker rats have abnormally low brain insulin content. Life Sci. 1985;36:627–633. doi: 10.1016/0024-3205(85)90166-3. [DOI] [PubMed] [Google Scholar]
  • [159].Israel PA, Park CR, Schwartz MW, Green PK, Sipols AJ, Woods SC, Porte D, Jr., Figlewicz DP. Effect of diet-induced obesity and experimental hyperinsulinemia on insulin uptake into CSF of the rat. Brain Res. Bull. 1993;30:571–575. doi: 10.1016/0361-9230(93)90084-o. [DOI] [PubMed] [Google Scholar]
  • [160].Kaiyala KJ, Prigeon RL, Kahn SE, Woods SC, Schwartz MW. Obesity induced by a high-fat diet is associated with reduced brain insulin transport in dogs. Diabetes. 2000;49:1525–1533. doi: 10.2337/diabetes.49.9.1525. [DOI] [PubMed] [Google Scholar]
  • [161].Kastin AJ, Akerstrom V, Pan W. Activation of urocortin transport into brain by leptin. Peptides. 2000;21:1811–1817. doi: 10.1016/s0196-9781(00)00349-1. [DOI] [PubMed] [Google Scholar]
  • [162].Pan W, Akerstrom V, Zhang J, Pejovic V, Kastin AJ. Modulation of feeding-related peptide/protein signals at the blood-brain barrier. J. Neurochem. 2004 doi: 10.1111/j.1471-4159.2004.02502.x. in press. [DOI] [PubMed] [Google Scholar]
  • [163].Blennow K, Wallin A, Fredman P, Karlsson I, Gottfries CG, Svennerholm L. Blood-brain barrier disturbance in patients with Alzheimer’s disease is related to vascular factors. Acta Neurol. Scand. 1990;81:323–326. doi: 10.1111/j.1600-0404.1990.tb01563.x. [DOI] [PubMed] [Google Scholar]
  • [164].de la Torre JC, Mussivand T. Can disturbed brain microcirculation cause Alzheimer’s disease? Neurological Research. 1993;15:146–153. doi: 10.1080/01616412.1993.11740127. [DOI] [PubMed] [Google Scholar]
  • [165].Kalaria RN. Neurodegenerative disease: Diabetes, microvascular pathology and Alzheimer disease. Nat Rev Neurol. 2009;5:305–306. doi: 10.1038/nrneurol.2009.72. [DOI] [PubMed] [Google Scholar]
  • [166].Kalaria RN. Cerebrovascular degeneration is related to amyloid-beta protein depositon in Alzheimer’s disease. Ann. NY Acad. Sci. 1997;826:263–271. doi: 10.1111/j.1749-6632.1997.tb48478.x. [DOI] [PubMed] [Google Scholar]
  • [167].Deloncle R, Guillard O. Mechanism of Alzheimer’s disease: arguments for a neurotransmitter-aluminium complex implication. Neurochem. Res. 1990;15:1239–1245. doi: 10.1007/BF01208586. [DOI] [PubMed] [Google Scholar]
  • [168].Grammas P, Botchlet T, Fugate R, Ball MJ, Roher AE. Alzheimer disease amyloid proteins inhibit brain endothelial cell proliferation in vitro. Dementia. 1995;6:126–130. doi: 10.1159/000106934. [DOI] [PubMed] [Google Scholar]
  • [169].Ikeda T, Furukawa Y, Mashimoto S, Takahashi K, Yamada M. Vitamin B12 levels in serum and cerebrospinal fluid of people with Alzheimer’s disease. Acta Psychiatr. Scand. 1990;82:327–329. doi: 10.1111/j.1600-0447.1990.tb01395.x. [DOI] [PubMed] [Google Scholar]
  • [170].Erkinjuntii T, Gauthier S. The concept of vascular cognitive impairment. Front Neurol Neurosci. 2009;24:79–85. doi: 10.1159/000197886. [DOI] [PubMed] [Google Scholar]
  • [171].Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M, Porte D., Jr. Cerebrosinal fluid and plasma insulin levels in Alzheimer’s disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology. 1998;50:164–168. doi: 10.1212/wnl.50.1.164. [DOI] [PubMed] [Google Scholar]
  • [172].Risberg J, Gustafson L. 133Xe cerebral blood flow in dementia and in neuropsychiatry research. In: Magistretti PL, editor. Functional Radionuclide Imaging of the Brain. Raven Press; New York: 1983. pp. 151–159. [Google Scholar]
  • [173].Arispe N, Rojas E, Pollard HB. Alzheimer disease amyloid β protein forms calcium channels in bilayer membranes: blockade by tromethamine and aluminum. Proc. Natl. Acad. Sci. USA. 1993;90:567–571. doi: 10.1073/pnas.90.2.567. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES