You are on page 1of 12

REvIEwS

The blood–brain barrier as an


endocrine tissue
William A. Banks
Abstract | The blood–brain barrier (BBB) was first noted for its ability to prevent the unregulated
exchange of substances between the blood and the central nervous system (CNS). Over time,
its characterization as an interface that enables regulated exchanges between the CNS and
substances that are carried in the blood in a hormone-​like fashion have emerged. Therefore,
communication between the CNS, BBB and peripheral tissues has many endocrine-​like
properties. In this Review, I examine the various ways in which the BBB exhibits endocrine-​
related properties. The BBB is a target for hormones, such as leptin and insulin, that affect many
of its functions. The BBB is also a secretory body, releasing substances either into the blood
or the interstitial fluid of the brain. The BBB selectively allows classical and non-​classical
hormones entry to and exit from the CNS, thus allowing the CNS to be both an endocrine
target and a secretory tissue. The BBB is affected by endocrine diseases such as diabetes
mellitus and can cause or participate in endocrine diseases, including those related to thyroid
hormones and obesity. The endocrine-​like mechanisms of the BBB can extend the definition
of endocrine disease to include neurodegenerative conditions, including Alzheimer disease,
and of hormones to include cytokines, triglycerides and fatty acids.

The blood–brain barrier (BBB) was originally defined These extra-​barrier properties create a blood–brain
by its ability to prevent the unregulated entry of blood-​ interface6 that is endowed with many endocrine-​like
borne substances into the central nervous system (CNS). properties (Fig. 2). In brief, these are the ability of brain
At least three barriers are currently recognized: a vas- endothelial cells to react to both traditional hormones
cular BBB formed by brain endothelial cells; a blood– and other circulating elements (thus, the barrier cells
cerebrospinal fluid (CSF) barrier formed by epithelial act as classic endocrine target tissues); the ability of
cells; and a barrier formed by tanycytes, which are special barrier cells, especially brain endothelial cells, to secrete
ependymal cells within the brain, that interfaces between substances into the blood that affect other cells (thus,
the circumventricular organs and other adjacent brain the barrier cells can act as classic endocrine secretory
tissue1. These barriers all possess tight junctions (Box 1) tissues); the ability of the BBBs to transport substances
that block intercellular leakage, have reduced macro­ from the blood into the brain and from the brain into
pinocytosis and contain no fenestrae (small pores in the the blood (thus, the BBBs selectively overcome their own
cell membrane), which together all reduce intracellular blockade, allowing the brain to act as an endocrine target
leakage (Fig. 1). and facilitating it as an endocrine secretory tissue); the
These BBBs perform many functions in addition to BBBs can be targets of endocrine diseases, and the result-
that of barrier formation. Through the transport of glu- ing alterations in BBB properties can underlie some of
cose, vitamins, minerals and other substances, the BBBs the clinical manifestations of those endocrine diseases;
Geriatric Research Education
supply the CNS with its nutritional needs2. Through and finally, malfunctions of the BBBs can cause endo-
and Clinical Center, Veterans
Affairs Puget Sound Health efflux transporters and enzymatic activities, the BBBs crine diseases. Together, these five properties provide
Care System and Division help rid the brain of toxins produced there and prevent unique ways in which the BBB can be involved in the
of Gerontology and Geriatric those exogenous to the CNS from accumulating or enter- treatment of endocrine diseases.
Medicine, Department ing the CNS in the first instance3. By regulating the influx In this Review, I explore these aspects of the endo-
of Medicine, University of
Washington School of
and efflux of informational molecules such as peptides crine BBB. Some of these properties have many exam-
Medicine, Seattle, WA, USA. and regulatory proteins, and because it is a source of ples, and I am unable to cover them all. Therefore, this
e-​mail: wabanks1@uw.edu informational molecules itself, the BBBs, and especially Review focuses on the endocrine-​like properties of the
https://doi.org/10.1038/ the vascular BBB, act as interfaces in a humoral-​based vascular BBB and defines the underlying principles that
s41574-019-0213-7 communication axis between the CNS and the blood4,5. are currently understood.

444 | AUGUST 2019 | volume 15 www.nature.com/nrendo


Reviews

Key points junctions, which, as mentioned earlier, are critical for


maintaining the barrier functions of the BBB. Most of
• The blood–brain barrier (BBB) acts as both a secretory and target endocrine tissue. these substances are secreted from cells on the brain
• By regulating the transport of hormones into and out of the brain, the BBB provides a side of the BBB, particularly pericytes and astrocytes.
mechanism by which the central nervous system can act as an endocrine secretory However, some classic hormones might also have a role
and target tissue. in regulating tight junctions, including glucagon-​like
• The BBB facilitates substances not typically thought of as hormones acting in an peptide 1 (GLP1) and transforming growth factor 1
endocrine-​like fashion, including triglycerides, short-​chain fatty acids and (refs31,32). In addition to substances that could regulate
lipopolysaccharide. tight junctions, substances that were not traditionally
• BBB function can be altered in endocrine diseases either because of adaptions to the thought of as hormones circulate in the blood and affect
disease condition or because it is a disease target. the function of the BBB and brain endothelial cells.
• BBB dysfunction or impairment can cause or promote the progression of endocrine or These include cytokines, triglycerides, free fatty acids
metabolic diseases. and lipopolysaccharide (LPS)32–35 (Box 2).
• The BBB is an important factor in the treatment of many diseases, often as a The aforementioned examples illustrate several prin-
therapeutic target or as a barrier that must be negotiated. ciples: barrier cells constitute an endocrine tissue that
responds to circulating factors; alterations in the BBB
affect all the known categories of BBB function (that is,
The BBB as an endocrine target effects on barrier properties, transporters, enzymatic
The many barrier and interface functions of the BBB activities and secretory processes); and some effects are
require its performance to be fine-​tuned to the needs mediated through classic hormones, such as insulin.
of the CNS and to be adaptable to the changing periph- Other blood-​borne effectors are not typically thought
eral environment. As such, the BBB responds to hor- of as hormones, such as triglycerides and LPSs, and
mones, which makes it an endocrine target tissue. For some effectors do not cross the intact BBB (for example,
example, oestradiol modulates sodium–potassium– adrenaline). Other effectors do cross, including those
chloride co-​t ransporters, and through this mech- with effects on other transporters. For example, insulin,
anism it can influence post-​stroke brain oedema7. which is transported across the BBB, increases leptin
1,25-dihydroxyvitamin D3 regulates the expression of the transport and triglycerides (which are also transported
efflux transporter LDL receptor-​related protein 1 (LRP1) across the BBB), affecting both insulin and leptin trans-
and the influx transporter receptor for advanced glyca- port. Thus, the scene is set for complex physiological
tion end products (RAGE)8 (of note, among the many lig- interactions. As a result of these endocrine effects, the
ands of these transporters are the amyloid-​β proteins9,10). BBB is able to adapt to environmental influences that,
Through the AT1 receptor, angiotensin II modi­fies BBB in turn, affect brain function. Hence, the BBB aids the
permeability, including via transcytosis11,12. Thus, angi- CNS in detecting and responding to environmental
otensin II could be the hormone responsible for opening conditions as presented to it through the bloodstream.
the BBB during hypertensive emergencies, resulting in Signals coming from the CNS to the brain endothelial
hypertensive encephalopathy13. cells allow the BBB to respond to the changing needs
Although insulin does not modulate GLUT1, which is of the brain.
the BBB transporter for glucose, insulin has other effects
on BBB functions. For example, insulin enhances the The BBB as an endocrine secretory tissue
transport of tryptophan across the BBB, which in turn All the barrier cells are able to secrete a number of sub-
could affect brain levels of serotonin and kynurenine, stances, but of the barrier cells, brain endothelial cells
substances that are related to sleep and depression14–16. are the most widely studied. Brain endothelial cells can
Insulin also regulates the activity or affects the levels of release nitric oxide, prostaglandins, cytokines and adre-
brain endothelial cell alkaline phosphatase, glutathione nomedullin36–41. The epithelial cells that constitute the
and P-​glycoprotein17–19, and insulin increases leptin trans- blood–CSF barrier and the tanycytes that form a bar-
port across the BBB20. The BBB transporter for insulin rier between circumventricular organs and adjacent
itself is, in turn, regulated by several substances, including brain tissue also have secretory functions42,43. Secretions
levels of iron, cholecystokinin and triglycerides21–23. are both constitutive and induced and are influenced
Many other functions of the BBB are regulated in an by the other cells of the neurovascular unit44. IL-6 and
endocrine-​like fashion. Leptin transport is modulated granulocyte–macrophage colony-​stimulating factor
by blood-​borne glucose, triglycerides, adrenaline and (GM-​CSF; which is classically know as a white blood
probably oestrogens20,24. Amylin alters tyrosine and tryp- cell growth factor) secreted from brain endothelial cells
Neurovascular unit tophan transport25, the BBB efflux pump peptide trans- act in an autocrine or paracrine fashion to enhance the
A multicellular and port system 1 (PTS1) is altered by branched-​chain amino translocation of the HIV-1 virus across the BBB 45.
multicomponent network that
acids26 and the mannose 6-phosphate receptor (which The secretions from barrier cells can also affect other
is composed of neurons, glial
cells, brain endothelial cells transports some lysosomal enzymes across the neonatal cells within the CNS. For example, brain endothelial cells
and extracellular matrix BBB) is modulated by circulating adrenaline and reti- secrete fever-​mediating prostaglandins46,47 and IL-1β
components. The neurovascular noic acid27–29. Evidence suggests that the decreases in the and IL-6, which influence the uptake of copper and iron
unit is key to neurovascular expression of GLUT1 associated with a high-​fat diet are by glial cells34,48.
coupling and to the delivery
of key nutrients and oxygen
rescued by vascular endothelial growth factor30. A mechanism for intercellular crosstalk within the
from the circulatory system A number of substances have been proposed that CNS, and the basis for an endocrine-​like axis that con-
into the brain. could regulate the induction or integrity of the tight nects peripheral tissue events with brain functions, is

NATURe RevIeWS | EndoCrinoLogy volume 15 | AUGUST 2019 | 445


Reviews

Box 1 | The endocrinology of the blood–brain barrier enter the brain is determined primarily by their lipid
solubility but is also influenced by their molecular mass
Both inside and outside the brain as well as the extent and type of plasma protein binding.
Brain endothelial cells define the limits of the central nervous system (CNS) and Accumulation within the brain is dictated by avidity of
therefore have a luminal membrane facing into the bloodstream and an abluminal the CNS binding receptor, especially when compared
membrane facing into brain interstitial fluid. Tight junctions cement adjoining brain
with the avidity of binding to plasma proteins. The
endothelial cells together, preventing intercellular diffusion (that is, blood–brain barrier
(BBB) leakage), but tight junctions also have a fence function154,155 — they block the presence of brain-​to-blood efflux systems can also affect
diffusion of proteins and lipids between the luminal and abluminal outer cell membrane the accumulation of a substance, as demonstrated for
leaflets. The tight junctions endow the BBB with characteristics of polarization; that is, dehydroepiandrosterone sulfate49.
the luminal surface membrane has very different transport, enzymatic and receptor As shown by the classic studies of Geraldo David
characteristics from the abluminal surface membrane. Such polarization underlies and Tapan Anand Kumar, and of Samuel Marynick and
many BBB characteristics, such as unidirectional transport or the ability to respond to a colleagues, the degree of uptake of steroid hormones
stimulus at one surface membrane by releasing a molecule from the other membrane. ranges widely, as exemplified by the accumulation of
The BBB is a barrier; the BBB is a gateway 17α hydroxyprogesterone being ~200 times greater
The BBB prevents the unrestricted entry of materials into the CNS. The lack of fenestrae than that of testosterone50,51. Many of the efflux systems
at the BBB, decreased macropinocytosis across the BBB and the presence of tight that include steroid hormones among their ligands are
junctions limit the way in which nutrients and other resources can enter the CNS. members of the organic anion transporting polypeptide
Therefore, the BBB meets the demand for nutrient transfer with a host of transporters, (OATP) and the ATP-​binding cassette (ABC) trans-
including those for glucose, amino acids, vitamins and minerals. porter families49,52,53. Non-​sulfated steroids are predom-
The BBB is constant; the BBB is ever changing inantly transported in the brain-​to-blood direction at
The robust barrier function of the BBB is consistent throughout the lifespan of the both the vascular BBB and the choroid plexus. However,
healthy individual. Contrary to popular misunderstanding, the fetal barrier is not leaky for hydrophilic sulfated steroid hormones, which are
to serum proteins, nor in most studies is the healthy aged BBB114,156. Similarly, rates of in far greater abundance than non-​sulfated steroids,
transmembrane diffusion are not different between the neonate and the adult157.
current evidence strongly suggests that the main direc-
However, transport rates of amino acids differ with development, and many transporters
are altered in aged individuals. These differences are because the BBB is slave to the
tion of transport is blood to brain and is mediated by
needs of the brain and therefore adapts to the changing needs of the brain throughout the OATPs53,54. Brain endothelial cells can also act as a
the lifespan — these changes can be adaptive or maladaptive and contributory enzymatic barrier to some steroids54.
to disease. Other classic hormones, such as thyroid hormones,
insulin, leptin and ghrelin, cross the BBB by saturable
processes. Many of the major saturable transport sys-
the process by which circulating substances act at the tems, such as GLUT1, are non-​energy-requiring systems
luminal side of brain endothelial cells to stimulate (that is, they function via facilitated diffusion). Other
the release of substances from these cells into brain inter- transport systems rely on pores or are transcytotic —
stitial fluid that then act on other brain cells. In sum- the latter always require energy and are variably classi­
mary, the ability of barrier cells to secrete substances into fied as carrier-​mediated transport, receptor-​mediated
blood establishes an endocrine-​like axis; the ability of transport, receptor-​mediated transcytosis and adsorp-
barrier cells to secrete substances into the brain com- tive transcytosis. A substance that is the primary ligand
partment in response to blood hormones establishes for a transport system typically crosses at a rate that is
a mechanism by which those hormones can influence 10–100-fold greater than the rate predicted on the basis
brain function without crossing the BBB. of its lipid solubility and other physicochemical charac-
The aforementioned examples illustrate several teristics2. A saturable transport system also means that
principles: all barrier cells (endothelial, epithelial and the BBB can negate the effect of binding to those plasma
tanycytic) are secretory; secretions can be constitutive proteins when the BBB transporter has a higher binding
or induced; for brain endothelial cells, secretions can affinity than the plasma protein.
be into the bloodstream or into the CNS; and paracrine
and autocrine effects have been demonstrated. Whether Thyroid hormones. Thyroid hormones are transported
substances derived and released by brain endothelial cells across cell membranes by various transporters, but only
into the bloodstream affect distal peripheral tissues in the monocarboxylate transporter 8 (MCT8) and OATP1C1
classic haemocrine fashion has not been demonstrated. are thought to be physiologically relevant55. Thyroid hor-
mone receptors do not function as BBB transporters, and
Transfer of hormones across the BBB the large neutral amino acid (LNAA) transporters LAT1
Pathways and mechanisms. The barrier function of and LAT2, although capable of transporting thyroid
the BBB would ordinarily deny water-​soluble hormones hormones, do not seem to have a physiological role.
Plasma protein binding entry to the CNS, thus making it impervious to many Evidence suggests that there are species specific dif-
The degree to which a
circulating signals. However, the BBB is permeable to ferences in the rate of thyroid hormone transfer across
substance binds to proteins
within the blood. many hormones, which makes the CNS an endocrine the BBB. For example, in humans thyroid hormone
target tissue. transfer relies on MCT8, whereas in murine endothelial
Choroid plexus There are many examples of circulating substances cells OATP1C1 is abundantly expressed in addition to
The choroid plexus, which that cross the BBB to act on the CNS. Classic exam- MCT8 (refs56,57). On the basis of mutation and knockout
consists of modified
ependymal cells, produces the
ples of substances that cross the BBB are the steroid studies, MCT8 in mice favours T3 transport, whereas
cerebrospinal fluid in the hormones that cross by the non-​saturable process of OATP1C1 favours T4 transport; MCT8 might also have
ventricles of the brain. transcellular diffusion. The rate at which these steroids some efflux function56. This combination of influx and

446 | AUGUST 2019 | volume 15 www.nature.com/nrendo


Reviews

a efflux transport is consistent with an early study in mice


Membrane protein
Nonpolarized cell finding that T3 crosses mainly in the blood-​to-brain
direction and T4 is dominated by an efflux system58.
Substrate Deiodinases are abundantly expressed in the brain59
and they are thought to have a major role not only in
Cytoplasm the conversion of T4 to T3 but also in the catabolism of
thyroid hormones to inactive forms. Finally, free iodide
is rapidly transported out of the brain60,61. MCT8 and
OATP1C1 are also abundantly expressed in the choroid
Substrate binding plexus. When injected into the lateral ventricle, however,
Cell membrane:
inner leaflet leads to response T3 and T4 penetrate only into the periventricular area.
Such limited periventricular distribution is the rule for
Nucleus
substances injected into the CSF62,63. As such, it is likely
that the choroid plexus is a source of thyroid hormones
Vesicle for brain regions that are in contact with the CSF.

Cell membrane: Insulin. Insulin is a classic hormone that in mammals is


outer leaflet secreted predominantly by a single tissue, the pancreas.
Insulin is transported across the BBB by a saturable
Exocytosis Endocytosis Extracellular fluid system64–66. In the periphery, insulin acts as a metabolic
hormone, inducing the uptake and metabolism of glu-
cose by tissues that express GLUT4, such as muscle, adi-
b pose tissues and the liver. Transport of glucose across
Brain Transcytosis Abluminal Abluminal the BBB, however, depends on the non-​insulin-sensitive
contents response GLUT1, and few brain cells possess GLUT4. Many of
the properties of CNS insulin are more mitogenic (that
Endothelial cell is, they are involved in growth) than metabolic (such
Tight Tight junctions
junction separate luminal as being used for energy); it seems as if peripheral and
and abluminal
contents CNS insulin have followed separate evolutionary paths67.
It should be noted, however, that studies that investi-
gate the actions of substances, not just hormones, on the
CNS often administer pharmacologically relevant doses,
Endocytosis Exocytosis therefore the elucidated actions on the CNS might not
Luminal Luminal
contents response
Activated occur physiologically. In addition, any substance that is
Blood receptor
injected into the CSF will eventually enter the blood-
stream as the CSF is reabsorbed. Studies have shown
Lipid that substances injected into the CSF can be found
Protein
peripherally in the blood. In some cases, those levels are
the same as if the substance had been given as an intra­
venous infusion68,69. Ironically, then, the effects produced
Abluminal outer after an intracerebroventricular injection can actually on
Abluminal membrane leaflet lipids occasion be mediated at peripheral sites of action.
membrane Research published between 1984 and 1992 reported
protein Inner membrane
leaflet lipid that CNS insulin influenced feeding and blood glucose
levels; however, the effects were paradoxical to those
Luminal Luminal outer of insulin-​induced hypoglycaemia70,71. That is, CNS
membrane membrane insulin inhibits feeding, elevates blood glucose and even
protein leaflet lipid
reduces blood insulin. Therefore, insulin is one of several
hormones that have paradoxical CNS effects, raising the
possibility that they act as their own counter-​regulatory
hormones72.
CNS insulin acts in the hypothalamus to control
Fig. 1 | The presence and architecture of tight junctions results in polarization ~50% of hepatic glucose production by stimulating
of the brain endothelial cell. a | Endocytosis, membrane protein distribution and sympathetic outflow to the liver73,74. Although, it is
receptor response in a nonpolarized cell all occur in a single extracellular environment. insulin’s mitogenic-​like effects on brain cells and its
b | Tight junctions create polarization so that the extracellular environment is divided possible use in the treatment of Alzheimer disease that
into luminal (blood) and abluminal (brain) spaces. Transcytosis (blood–brain barrier
have raised the most excitement of late75. As mentioned
penetration) and exocytosis can occur. Receptor activation can result in a response on
the luminal or abluminal side of the cell. Inset shows that the fence function of tight previously, the actions of insulin in the CNS are more
junctions prevents the exchange of luminal and abluminal outer membrane leaflet mitogenic than metabolic and resemble more closely
contents, allowing for unique populations of lipids and proteins. The fence function those of the ancestral insulins than those of peripheral
extends only to the outer leaflet and therefore does not divide the inner leaflet into insulin76. Insulin administered to the brains of healthy
distinct luminal and abluminal regions. humans, cognitively impaired humans or mouse models

NATURe RevIeWS | EndoCrinoLogy volume 15 | AUGUST 2019 | 447


Reviews

Brain Blood-to-brain passage


of hormones across the
BBB allows the brain to
be an endocrine target

BBB

Receptor

The BBB is a target for The BBB secretes Autocrine Paracrine Blood-to-brain
circulating substances substances into the effects effects passage of
secreted by other tissues circulation that substances across
affect other tissues the BBB allows the
brain to act as an
endocrine
secretory tissue

Blood

Fig. 2 | The vascular blood–brain barrier as an endocrine tissue. The vascular blood–brain barrier (BBB) is a target
for circulating substances in the blood7,8,11,12,17–19. In addition to this, the vascular BBB secretes substances into the blood
that act in autocrine, paracrine and haemocrine fashions34,36–41,45–48 and it regulates the exchange of informational
substances between the central nervous system and the blood50,51,55,64–66,80,81, resulting in humorally mediated brain–body
communication.

of Alzheimer disease results in rapid improvement in The glucose-​n ormalizing effect of FGF1 is not
cognition and has effects on neuronal survival, plasticity dependent on weight loss, reduced food intake, changes
of synapses, dendritic arborization and the formation of in basal glucose turnover or production rate, glucose
neuronal circuits77–79. uptake by heart or adipose tissues or changes in blood
insulin or glucagon levels. Unlike FGF15/FGF19, FGF1
Ghrelin. Ghrelin is illustrative of other hormones has little or no effect on the hypothalamic–pituitary–
that are predominantly synthesized in the periphery adrenal axis and only has acute effects on sympa-
but are capable of crossing the BBB80,81 in quantities thetic outflow92. Instead, FGF1 administered by intra­
sufficient to influence CNS function. Aside from having cerebroventricular injection was shown to increase
effects on feeding and energy expenditure, ghrelin also hepatic and skeletal muscle uptake of glucose, a process
has effects on cognition, synaptic function, neuronal that probably occurred via an insulin receptor-​dependent
survival and neurogenesis (effects that are shared by mechanism92.
insulin and leptin)82–85.
Leptin. The examples of BBB–hormone transport path-
Fibroblast growth factors. Fibroblast growth factors ways that I have provided in this Review so far mostly
(FGFs) are a large family of regulatory proteins with relate to brain endothelial cells and the vascular BBB. It is
many functions, including endocrine functions. FGF2 worth noting, however, that although the epithelial bar-
(basic FGF), FGF19 (human homologue of mouse rier cells of the choroid plexus and tanycytes found at the
FGF15, henceforth referred to as FGF15/FGF19) and circumventricular organs share characteristics with brain
FGF21 have all been shown to cross the BBB, and there endothelial cells and the vascular BBB, each barrier results
is preliminary evidence for FGF1 (acidic FGF) cross- in unique distribution patterns for the hormones or other
ing86–89. In many cases, the ability of a peripherally substances it transports93,94. The unique regional distri-
administered FGF to cross the BBB underlies its ability in bution patterns are a result of the very low diffusion rates
part or in whole to induce CNS effects, including those within brain tissue. Furthermore, the low diffusion
on analgesia86, circadian behaviour90 and metabolism91. rates from the CSF into brain tissue means that when a
FGF15/FGF19 and FGF21 can circulate as hormones substance is introduced into the CSF it is fairly evenly dis-
and have acute metabolic effects that are mediated, at tributed throughout cranial CSF, but diffusion from the
least in part, through the liver and adipose tissues91. Both CSF into adjacent brain tissue is poor. As a result of this
FGF15/FGF19 and FGF21 increase energy expenditure poor diffusion, a substance transported into the CSF by
and decrease plasma insulin, glucose and triglyceride the choroid plexus remains mostly periventricular60,62,63,95.
concentrations in the liver in obese animals, but in a In theory, the three barriers (the vascular BBB, the
chronic setting these effects, as well as weight loss, are blood–CSF barrier and the tanycytic barrier at the cir-
predominantly mediated through the CNS. Part of the cumventricular organs) work in harmony to support
CNS effect of FGF15/FGF19 and FGF21 is mediated brain function, but there are some examples of how these
through sympathetic outflow from the CNS to brown barriers would affect a specific hormone or influence a
fat. Peripherally administered FGF1 also acts within the given brain region (Box 3; Fig. 3). Studies in rodents that
CNS to decrease hyperglycaemia92. investigated how peripheral leptin is distributed in the

448 | AUGUST 2019 | volume 15 www.nature.com/nrendo


Reviews

Box 2 | The blood–brain interface expands the definition of hormones Dramatic changes occur in these systems with food
restriction. After 24 h of fasting, leptin transport across
Hormones were originally defined as substances secreted by glandular tissues, but the the vascular BBB increases98, the tanycytic barrier extends
definition has long been extended to include secretions from non-​glandular tissues. further along the ventricle and some of the brain endo­
Some substances not traditionally thought of as hormones interact with the blood–brain thelial cells in the arcuate nucleus closest to the median
barrier (BBB) in an endocrine-​like fashion.
eminence lose barrier function93. Whereas these events
Cytokines probably increase leptin levels in the arcuate nucleus
Cytokines are transported across the vascular BBB and are secreted by brain endothelial after 24 h of fasting, the changes at 48 h act in unison to
cells and the epithelial cells of the blood–cerebrospinal fluid (CSF) barrier. With regard decrease leptin action, resulting in leptin deficiency, leptin
to the BBB, cytokines have autocrine, paracrine and haemocrine modes of action. The
peripheral resistance and leptin central resistance. By 48 h
endocrine-​like interactions between the BBB and cytokines are a major component of
the neuroimmune axis and participate in the central nervous system (CNS)-mediated
of fasting, serum leptin levels are decreased, the leptin
effects of circulating cytokines, including feeding, sleep, thermogenesis and fever, pain transport rate across the vascular BBB is greatly decreased
perception and the progression of neurodegenerative diseases104. and there is resistance at the leptin receptor. The latter
two events — inhibition of leptin transport and receptor
Triglycerides, free fatty acids and short-​chain fatty acids
Triglycerides act at the BBB to regulate the transport of leptin, ghrelin and insulin. They
binding — are mediated at least in part by triglycerides,
also cross the BBB to induce resistance to leptin and insulin at their CNS receptors. Free which are greatly increased in the blood with starvation
fatty acids, such as palmitate and arachidonate, cross the BBB, where they participate and are transported across the BBB33,99 (Fig. 3).
in membrane turnover and signalling. Free fatty acids act at the hypothalamus to affect The CNS can contribute levels of a substance in the
the expression of feeding hormones such as galanin and orexin. Short-​chain fatty acids blood. Such substances enter the bloodstream not only
(such as acetate, propionate and butyrate) connect the microbiome to the brain. through efflux transporters located at the vascular BBB
Gut bacteria act on dietary fibre to produce these short-​chain fatty acids, which enter and choroid plexus but also with the reabsorption of CSF
the blood and cross the BBB to affect brain functions. into the bloodstream. Indeed, because of this efflux, sub-
Lipopolysaccharides stances injected into the lateral ventricles of the brain
Lipopolysaccharides (LPSs) have potent effects on the BBB. For example, they can alter can produce blood levels that resemble that of an intra-
the function of BBB transporters; induce the release of prostaglandins, nitric oxide and venous infusion68. The renowned neuroendocrinologist
cytokines from the BBB; and disrupt the BBB104. LPSs can translocate across the gut Seymour Reichlin showed that on stimulation, the CNS
after physiological events, such as intense running, and circulate in blood under normal can be an impressive source of blood cytokines100–102.
conditions. The presence of LPSs in the blood in physiological conditions raises the
Others have shown that corticotropin-​releasing hormone
possibility of them having physiological roles, which might be mediated through their
given into the lateral ventricle of the brain can enter the
effects on the BBB; however, as colonizing bacteria are the source of LPSs, rather than
the human host, LPSs would be better described as a vitamin, albeit one with hormonal circulation in amounts sufficient to modulate splenic
effects, analogous to vitamin D. secretion of IL-1β100.

The microbiota. Once in the bloodstream, the secreted


arcuate nucleus following transport across the BBB in a products of the gut microbiota might have many effects
fed and non-​fed state offer some interesting examples on BBB functions and the neuroimmune axes in which
of how the three barriers can interact (Fig. 3). In the fed the BBB participates103,104. In addition, the products
state, leptin is transported across the vascular BBB by of the microbiome, as exemplified by the short-​chain
brain endothelial cells at the rate of ~5.9 × 10−4 ml/g, or fatty acids, cross the BBB to exert effects on the CNS.
~20 times faster than albumin80. In addition to the vas- A 2016 study showed that short-​chain fatty acids cross-
cular BBB, a second source of leptin is transported across ing the BBB increase microglial activation to the extent
the choroid plexus and into the CSF69,96. that the nature of the microbiome could be a risk factor
As mentioned earlier, owing to low diffusion rates for Parkinson disease105.
from the CSF to the brain, tissue concentrations of
BBB-​t ransported leptin decrease dramatically and The brain as an endocrine tissue. As detailed in this
logarithmically from the ventricular surface63 so that section, a number of fundamental principles govern
leptin remains mainly in the periventricular region of the transfer of hormones across the BBB. First, every
the arcuate nucleus69. The study by Berislav Zlokovic major class of hormone (steroids, peptides, regulatory
and colleagues96, which was conducted in rats, reported proteins and thyroid hormones) has representatives that
a transport rate at the choroid plexus that was ten times cross the BBB at physiologically relevant levels. Second,
higher than the transport rate found by Naoko Nonaka a variety of transfer mechanisms across the BBB exist,
and colleagues97 in the mouse. One explanation could be with transcellular diffusion and blood-​to-brain satura-
that the rat, with its larger arcuate nucleus and therefore ble transporters being the best understood mechanisms.
its larger diffusional distances, requires higher rates of Third, for some hormones, brain-​to-blood transporters,
leptin transport into the CSF than the mouse. plasma protein binding, enzymatic barrier function or
A third pathway that leptin can take to the arcuate CNS receptor affinity is a major or the major determi-
nucleus tissue is via diffusion across the leaky BBB of the nant of CNS uptake or retention. Furthermore, in many
median eminence. Leptin that enters the CNS via this cases, the protein acting as the hormone’s receptor is not
route is subsequently transported by tanycytes into the the same protein acting as its transporter.
CSF and arcuate nucleus tissue. Without this transport This section elucidated several other points. As illus-
function, the barrier function of the tanycytes would trated by leptin, the various barrier systems (endothelial–
prevent leptin from diffusing from the median eminence vascular, epithelial–CSF and tanycytic–CSF–circum-
into the CSF or arcuate nucleus93. ventricular organ) have ‘spheres of influence’; that is,

NATURe RevIeWS | EndoCrinoLogy volume 15 | AUGUST 2019 | 449


Reviews

Allan–Herndon–Dudley
the distribution of a hormone within the CNS differs gut microbiome such as short-​chain fatty acids can cross
syndrome depending on the barrier it has crossed. Leptin is also the BBB to influence the CNS.
A condition resulting from an example of how BBB–hormone transporters are not
deficient thyroid hormone static but are regulated by peripheral events, including The BBB as a target of endocrine disease
transport across the blood–
brain barrier characterized
by other hormones. Both the BBB and the blood–retinal barrier are disrupted
by cognitive impairment, lack In some cases, the effect of a hormone on the CNS in diabetes mellitus in both humans and animal models
of speech and hypotonia. is complementary to its peripheral action (CNS insu- of diabetes106,107. In streptozotocin-​induced diabetes, BBB
lin’s effect on hepatic glucose production); in other disruption is progressive, involving bigger lesions and
cases, it can be contradictory so that in the CNS the more regions of the brain with disease duration107. Blood–
hormone acts in counter-​regulatory fashion to its retinal barrier disruption, which results in diabetic
peripheral effects (CNS insulin’s ability to increase retinopathy, is the leading cause of blindness in Western
blood sugar and suppress appetite). In addition to countries. BBB disruption also occurs in diabetes
these outcomes, the effects induced in the CNS by a mellitus. Disruption in the occipital cortex can contrib-
hormone can seem unrelated to those of its peripheral ute to the visual deterioration108 seen in diabetes. BBB
actions (CNS insulin’s effect on cognition). As with disruption is closely linked with cognitive impairments109
the other endocrine functions of the BBB, substances and precedes cognitive impairment110.
acting in a hormone-​like fashion include those not In both BBB and blood–retinal barrier disruption, the
typically classified as hormones and have actions that main cause of disruption seems to arise from pericytic
are not typically thought of as endocrine (triglycerides glucotoxicity111,112. Pericytes, which are instrumental in
and leptin resistances). Furthermore, a blood-​borne instructing brain endothelial cells to form and maintain
hormone might exert an effect in the periphery that barrier functions113,114, take up glucose in an insulin-​
is dependent on a CNS site of action (insulin’s effect independent manner so that at high levels more glucose
on CNS-​mediated hepatic glucose output). In this enters the tricarboxylic acid (TCA) cycle, producing
case, the blood-​to-brain transfer of a hormone and more ATP but also increasing oxidative stress. This oxi-
the subsequent CNS action is part of a more complex dative stress is blamed for pericyte death, which leads
brain–body interaction. to BBB disruption and blood–retinal barrier disruption.
Research has also shown that brain-​to-blood transfer Pericyte loss has been proposed to have a role in other
mechanisms (such as transporters and CSF reabsorp- types of brain diseases with a BBB component, including
tion) can, under some circumstances, contribute mark- Alzheimer disease, cerebral cavernous malformation and
edly to blood levels of a hormone as illustrated by leptin, methamphetamine toxicity115–117.
cytokines and potassium. In addition, products from the In addition to pericyte integrity, other aspects of
the BBB can be affected by diabetes mellitus or obesity.
Leptin, choline and insulin transport, as well as the efflux
Box 3 | Three barriers and their spheres of influence transporter LRP1, are decreased in diabetes mellitus or
This Review has emphasized the vascular blood–brain barrier (BBB). This barrier is the with obesity98,118–121. Furthermore, the expression of
best studied of the three main barriers and fits most readily into an endocrine model numerous proteins by brain endothelial cells, such as
given that hormones are by definition blood-​borne. It is of note, however, that two cytoskeletal proteins, enzymes and transport-​related
other arms of the BBB, which are composed of epithelial cells and tanycytes, also have proteins, is decreased with obesity122. The lipid com-
endocrine-​like functions. In addition, areas of the brain in which capillaries lack barrier position of brain endothelial cells is not altered in the
functions (known as the circumventricular organs) have access to hormones much the brains of patients with diabetes mellitus or obesity, but
way peripheral tissues do. The circumventricular organs are connected to other regions brain endothelial cells in the brains of these individuals
of the brain by afferent and efferent nerves so that signals received in these non-​barrier
do have increased oxidative damage123.
regions of the brain can be relayed to other regions.
Hormones that enter the circumventricular organs are not able to diffuse to adjacent
Hyperhomocysteinaemia is also associated with BBB
brain regions nor to the cerebrospinal fluid (CSF) because of a delimiting tanycytic disruption124. Evidence suggests that disruption is via the
barrier (Fig. 3). The cells that form the tanycytic and epithelial barriers have transporter N-​methyl-d-​aspartate receptor, a finding that is consist-
functions and the epithelial cells of the choroid plexus have secretory properties ent with homocysteine being an agonist for this receptor.
as well158,159. As for diabetes mellitus, BBB disruption precedes the
The distribution of a hormone within the central nervous system (CNS) is greatly cerebral pathology found in this condition125.
influenced by which barrier a hormone has used to enter the CNS, creating unique In summary, barrier and transporter functions can
spheres of influence for each barrier. For example, hormones that leak into the space be altered in the brains of patients with endocrine and
between the capillary plexus and epithelial cells of the choroid plexus can influence meta­bolic diseases. Furthermore, barrier dysfunction
epithelial cell function or be transported into the CSF. Hormones that leak into
leads to clinical disease, as exemplified by blindness
the circumventricular organs, aside from influencing the brain cells within the
circumventricular organs, also have the potential of influencing the delimiting
resulting from blood–retinal disruption and cognitive
tanycytes or of being transported into adjacent brain tissue or CSF. Substances changes resulting from BBB disruption.
that enter the CSF have very limited diffusion into the brain and are mainly in the
periventricular and other CSF-​contacting areas. Likewise, substances diffusing The BBB as a cause of endocrine disease
across the tanycytic barrier are mainly confined to the adjacent tissue. Thyroid-​ h ormone-related conditions. Impaired
The vascular BBB could homogeneously distribute a hormone throughout the brain, expression of MCT8, the main BBB transporter of
but most hormones that cross the vascular BBB demonstrate a heterogeneous pattern thyroid hormones in humans, results in Allan–Herndon–
because of differences in transport, efflux and degradation rates. A fourth pattern of Dudley syndrome55. Brain lesions are present prenatally.
distribution occurs for hormones or hormone analogues that have very long half-​lives Interestingly, mice with an MCT8 deficiency do not
in blood as they can enter using the extracellular pathways160.
show altered brain structures or evidence for cognitive

450 | AUGUST 2019 | volume 15 www.nature.com/nrendo


Reviews

a Fed state b 24 h fasted c 48 h fasted


Central insulin
CSF Choroid plexus resistance
Arcuate nucleus Arcuate nucleus
Arcuate
nucleus
cell

Leptin
Leptin Triglyceride
receptor

Capillary ↓ Leptin
↑ Triglyceride
Ventromedial region Ventromedial region levels

Epithelial cell Leaky

Ependymal
cell

Tanycyte

Leaky
Median eminence

Fig. 3 | Blood-​to-brain transport of leptin into the arcuate nucleus: integration of signals and changes with feeding
state. a | In the fed state, blood-​derived leptin (blue dots) reaches the arcuate nucleus by transport across local capillaries
with barrier function80, transport across the epithelial cells of the choroid plexus into cerebrospinal fluid (CSF) with
subsequent leakage into brain tissue across leaky ependymal cells69,96 and transport into CSF and adjacent brain tissue by
the tanycytic barrier after leakage into the median eminence93. b | Three changes found at 24 h of fasting are an extension
of the tanycytic barrier into the ventricular region, an increase in the blood–brain barrier (BBB) transport rate of leptin
and the conversion of some capillaries in the ventromedial region of the arcuate nucleus to a leaky state93,98. c | At 48 h of
fasting, leptin blood levels are greatly reduced, blood triglycerides are elevated, blood triglycerides impair leptin transport
across the BBB contributing to peripheral leptin resistance and triglycerides cross the BBB to interfere with activation
of the leptin receptor, contributing to central leptin resistance33,93,98,99.

impairments. This lack of pathology in MCT8-deficient in maternal iodine deficiency126. Cretinism and MCT8
mice is thought to be caused by the higher expression deficiency have many differences in disease course; for
of OATP1C1 in mice56,57. OATP1C1 favours the trans- example, cretinism results in maternal and fetal whole-​
port of T4, but high levels of deiodinases in brain con- body thyroid hormone deficiency, whereas MCT8
vert T4 to T3. Mice lacking both MCT8 and OATP1C1 deficiency has a paradoxical stage during the perinatal
have delayed cerebellar development, reduced mye- period of cerebral hyperthyroidism. Cerebral hyper­
lination and other pathologies consistent with CNS thyroidism does not occur in MCT8-deficient mice that
hypothyroidism56. also have a knockout of LAT2 (ref.127), suggesting that
Interestingly, in humans, MCT8 deficiency results LAT2 might be an important transporter of T3 in neo-
in a very different clinical presentation from cretin- nates and might even be upregulated during this period
ism, another form of thyroid deficiency that has roots in the absence of MCT8.

NATURe RevIeWS | EndoCrinoLogy volume 15 | AUGUST 2019 | 451


Reviews

Box 4 | What controls feedback outside a negative feedback loop? downstream of the leptin receptor138–140. Resistance at
the BBB occurs early with the onset of obesity and likely
The concept of the negative feedback loop is central to classic endocrinology. Such a participates in a positive feedback-​loop-like fashion to
loop results in the fine-​tuning of the levels of the effector hormone and the end point. promote obesity.
A classic example of a feedback loop is the relationship between glucose and insulin. The mechanisms underlying the development, pro-
Insulin levels in blood increase when blood glucose levels rise, which results in glucose
gression and maintenance of obesity and leptin resist-
levels returning to within the normal range. With the decrease in blood glucose levels,
the stimulus for insulin secretion is abated and blood insulin levels also return to ance have been influenced by evolutionary pressures to
previous values. acquire excess calories and to retain them as fat. Wild-​
Insulin has actions within the central nervous system (CNS), and there must also be living adult primates have fat masses of ~6% of their
an optimal level of brain insulin. The actions of insulin in the brain are more mitogenic body weight141, which is about the same as seen in per-
than metabolic and are not coupled to its effects on blood glucose, thus there is a sons with anorexia nervosa142. Hunter-​gatherers typically
dissociation in the control of end points. In other words, the optimization of blood have BMIs of ~19 (ref.143). In other words, the evolution-
insulin levels for the control of blood glucose does not ensure that brain insulin levels ary struggle has been to acquire a caloric reserve to see
are optimized for its CNS functions. This theoretical dilemma exists for other hormones the individual through times when caloric acquisition
that are totally, or mainly, produced in the periphery but have actions in the CNS falls short of immediate requirements and not to avoid
that are not connected to their peripheral feedback mechanisms, including leptin
obesity and its metabolic consequences.
and ghrelin.
These evolutionary pressures can be seen in the BBB
pharmacokinetic curves for leptin80,134. Leptin transport
Glucose and insulin. Impaired transport of glucose across the BBB is saturable, which means that the rela-
and insulin across the BBB have each been proposed tionship between serum and CNS leptin levels resem-
as important factors in Alzheimer disease128. Impaired bles a hyperbolic curve. Where this hyperbolic curve is
transport of glucose across the BBB certainly results in linear, the blood concentrations of leptin are such that
cognitive impairments, seizures and ketosis as exem- they produce the greatest proportional increases in CNS
plified by the GLUT1 deficiency syndrome129. Despite leptin levels. As blood leptin levels increase, the curve
having an elevated serum insulin level, patients with flattens (loses linearity), which results in a smaller pro-
Alzheimer disease have a decreased level of insulin portion of blood leptin entering the CNS134,144. This loss
in the CSF as well as a decreased CSF:serum ratio for of linearity is the pharmacokinetic proximal cause and
insulin, suggestive of a defect in the BBB transport an immediate cause of peripheral resistance to leptin.
of insulin130. Insulin resistance in the CNS, so-​called The loss of linearity for both rodent brain levels of leptin
type 3 diabetes mellitus, can occur independently of and human CSF levels of leptin occurs between 5 and
peripheral insulin resistance131; indeed, the relationship 10 ng/ml (refs134,145), a level typically associated with
and interactions between these two spheres of insulin ideal body weight146. As such, leptin resistance is already
resistance are largely unexplored (Box 4). Tine Sartorius occurring in humans at levels of BMI that Western med-
and colleagues132 have noted that in comparison with icine has traditionally labelled ideal body weight, but not
young mice, aged mice have a delayed increase in cor- at levels seen in wild-​living primates or hunter-​gatherers.
tical brain activity after the peripheral, but not after the Thus, saturable transport at the BBB of a hormone
central, administration of insulin, suggesting that an adds an additional level of potential resistance to that
impaired transport of insulin across the BBB could be of a classic resistance syndrome. In non-​barrier tissues,
a contributor to CNS insulin resistance132. receptor resistance can be addressed by increased pro-
duction of the hormone until either the response returns
Leptin and leptin resistance. The BBB is directly invol­ to normal or hormone production reaches its max-
ved in peripheral leptin resistance and is indirectly imum. However, when the hormone crosses the BBB
involved in central leptin resistance, thus contributing by a saturable mechanism, the BBB can act as a choke
to obesity, the metabolic syndrome and diabetes mellitus. point. Saturation of the transporter at the BBB limits
Leptin is secreted primarily by adipose cells and is trans- how much hormone can enter the brain, and that limit
ported across the BBB by a saturable system80,133. Leptin’s can be insufficient to raise brain tissue levels to those
rate of transport into the arcuate nucleus area of the needed to overcome receptor resistance. In the case of
hypothalamus is particularly high but is also robust into leptin, peripheral resistance is the term used to denote
the hippocampus134. A loss of the anoretic effects of leptin the inability of the BBB to deliver sufficient amounts
results in profound obesity in both rodents and humans of hormone to the brain to overcome brain receptor
— leptin-​deficient mice at 10 months of age weigh more (central) resistance.
than three times their leptin-​replete littermates, and Triglycerides are a cause of peripheral leptin resist-
leptin-​deficient humans have BMIs in the low 50s135,136. ance, and elevation of triglycerides is the classic dyslipi-
Leptin-​deficient humans and animals are exqui- daemia of the metabolic syndrome. Triglycerides (Fig. 3)
sitely sensitive to treatment with leptin. For example, 18 act at the brain endothelial cell barrier to decrease the
months of leptin replacement therapy resulted in BMI rate of leptin transport across the BBB, thus they are
levels decreasing from an average of 51.2 to 26.9 (ref.135). capable of contributing to peripheral leptin resistance33.
Leptin deficiency is rare in humans, with cohorts of Triglycerides are also transported across the BBB and
individuals in Turkey135 and Paraguay137. Elevated blood can act at the CNS leptin receptor to induce central
leptin levels in the face of obesity are common in humans leptin resistance99. Thus, the BBB as either a target or
and can result from attenuated transport at the BBB, a conduit is involved in both peripheral and central
resistance at the leptin receptor or deficits in signalling leptin resistance.

452 | AUGUST 2019 | volume 15 www.nature.com/nrendo


Reviews

BBB transporter dysfunction and endocrine disease. Diabetes mellitus and insulin resistance. The poten-
In summary, BBB transporter dysfunction occurs for a tial merit of the delivery of FGFs to the brain for the
wide range of substances including thyroid hormones, treatment of diabetes mellitus is clear from the work dis-
glucose, insulin and leptin. Dysfunction of BBB trans- cussed above. Indeed, the discovery92 that when doses
porters results in changes in a wide range of symptoms, of FGFs one-​tenth of an effective peripheral dose are
including cerebral hypothyroidism, cognitive impair- delivered into the brain they reverse (apparently per-
ment, metabolic dysfunctions and obesity. To date, the manently) mild to moderate levels of hyperglycaemia in
clearest connections between BBB transporters and dis- some models of moderate obesity shows the potential of
ease exist for substances the brain does not synthesize combining the endocrine brain and the endocrine BBB.
or can synthesize only in limited amounts. The involve- Modifications to transporters, theoretically exemplified
ment of the BBB in an endocrine disease might be due by the ability of α-​adrenergics to overcome peripheral
to the primary loss of a transporter, as exemplified by leptin resistance or triglycerides to enhance transport
Allan–Herndon–Dudley syndrome, or as a downstream of insulin and ghrelin, could enable increased access of
event of the disease or condition that modulates the endogenous hormones to the CNS23,33,150.
transporter, as exemplified by hypertriglyceridaemia Peripheral insulin resistance, impaired transport of
and the metabolic syndrome. glucose and insulin across the BBB and central insulin
resistance are all implicated in Alzheimer disease. Dual
Endocrine BBB and treatment of diseases incretin receptor agonists (twincretins) directed at GLP1
The multiple facets of the endocrine BBB that make it and glucose-​dependent insulinotropic polypeptide that
vulnerable to being a target or a cause of endocrine dis- can cross the BBB are able to improve peripheral glucose
ease also suggest roles for it in the treatment of endocrine control and improve the BBB transport of glucose151–153.
diseases. Clearly, the role of the BBB as a transporter of Whether they can affect CNS insulin resistance or affect
hormones into the CNS, which allows the CNS to act as insulin transport across the BBB is yet to be published.
an endocrine target tissue, presents many possibilities. GLP1 has also been proposed to prevent diabetic disrup-
tion of the BBB by limiting the oxidative stress that arises
Phenylketonuria. Phenylketonuria results from a defec- from the hyperglycaemic conditions31.
tive metabolism of the LNAA phenylalanine. The other
LNAAs share with phenylalanine a transporter across Conclusions
the BBB, therefore an excess of circulating phenylalanine The BBB performs as both an endocrine target and an
results in both an increase in brain phenylalanine and a endocrine secretory tissue. By selectively regulating the
decrease in brain levels of other LNAAs caused by com- exchange between the CNS and the blood of traditional
petitive inhibition147. CNS symptoms can arise both as hormones as well as other informational molecules, the
a result of toxic levels of phenylalanine and because of a BBB also allows the CNS to act as both an endocrine tar-
deficiency in neurotransmitter substrates. Although the get and an endocrine secretory tissue. The BBB is a target
disease is typically treated with diets low in phenylala- for some endocrine diseases and is an active participant
nine, several studies have advocated treating with a mix- in the onset and promulgation of endocrine disease; as a
ture of LNAAs, which would both block phenylalanine result, the BBB can also be key to the treatment of some
from crossing the BBB in high amounts and increase the endocrine diseases. Finally, the view of the BBB as an
uptake of LNAAs that act as precursors to neurotrans- endocrine tissue expands the list of hormones to include
mitters148,149. A similar strategy has also been advocated cytokines, lipids and even LPSs.
for maple syrup urine disease, in which branched-​chain
LNAAs accumulate in the brain148. Published online 24 May 2019

1. Neuwelt, E. et al. Strategies to advance translational enhancing Abeta1 = 40 brain-​to-blood efflux and into the brain of the rabbit. J. Physiol. 312, 551–562
research into brain barriers. Lancet Neurol. 7, 84–96 peripheral uptake transport. Neruoscience 322, (1981).
(2008). 28–38 (2016). 16. Walker, A. K., Wing, E. E., Banks, W. A. & Dantzer, R.
2. Davson, H. & Segal, M. B. (eds) in Physiology of the 9. Deane, R., Wu, Z. & Zlokovic, B. V. RAGE (yin) versus Leucine competes with kynurenine for blood-​to-brain
CSF and Blood–Brain Barriers 303–485 (CRC Press, LRP (yang) balance regulates Alzheimer amyloid transport and prevents lipopolysaccharide-​induced
Boca Raton, 1996) beta-​peptide clearance through transport across the depression-​like behavior in mice. Mol. Psychiatry.
3. Banks, W. A. in Efflux Transporters and the Blood-​Brain blood-​brain barrier. Stroke 35, 2628–2631 (2004). https://doi.org/10.1038/s41380-018-0076-7 (2018).
Barrier (ed. Taylor, E. M.) 21–53 (Nova Science 10. Deane, R., Sagare, A. & Zlokovic, B. The role of the cell 17. Langston, J. W., Li, W., Harrison, L. & Aw, T. Y.
Publishers Inc.,2005). surface LRP and soluble LRP in blood-​brain barrier Activation of promoter activity of the catalytic subunit
4. Pan, W. & Kastin, A. J. Cytokine transport across the A beta clearance in Alzheimer’s disease. Curr. Pharm. of gamma-​glutamylcysteine ligase (GCL) in brain
injured blood-​spinal cord barrier. Curr. Pharm. Des. Des. 14, 1601–1605 (2008). endothelial cells by insulin requires antioxidant
14, 1620–1624 (2008). 11. Fleegal-​DeMotta, M. A., Dohgu, S. & Banks, W. A. response element 4 and altered glycemic status:
5. Kastin, A. J. & Pan, W. Blood-​brain barrier and Angiotensin II modulates BBB permeability via implication for GCL expression and GSH synthesis.
feeding: Regulatory roles of saturable transport activation of the AT1 receptor in brain endothelial Free Radic. Biol. Med. 51, 1749–1757 (2011).
systems for ingestive peptides. Curr. Pharm. Des. 14, cells. J. Cereb. Blood Flow Metab. 29, 640–647 18. Liu, H. et al. Insulin regulates P-​glycoprotein in rat brain
1615–1619 (2008). (2009). microvessel endothelial cells via an insulin receptor-
6. Banks, W. A. From blood-​brain barrier to blood-​brain 12. Guillot, F. L. & Audus, K. L. Angiotensin peptide ​mediated PKC/NF-​kappaB pathway but not a PI3K/Akt
interface: new opportunities for CNS drug delivery. regulation of fluid-​phase endocytosis in brain pathway. Eur. J. Pharmacol. 602, 277–282 (2009).
Nat. Rev. Drug Discov. 15, 275–292 (2016). microvessel endothelial cell monolayers. J. Cereb. 19. Catalan, R. E., Martinez, A. M., Aragones, M. D.,
7. O’Donnell, M. E., Lam, T. I., Tran, L. Q., Foroutan, S. Blood Flow Metab. 10, 827–834 (1990). Miguel, B. G. & Robles, A. Insulin action on brain
& Anderson, S. E. Estradiol reduces activity of the 13. Vaughan, C. J. & Delanty, N. Hypertensive emergencies. microvessels; effect on alkaline phosphatase. Biochem.
blood-​brain barrier Na-​K-Cl cotransporter and Lancet 356, 411–417 (2000). Biophys. Res. Commun. 150, 583–590 (1988).
decreases edema formation in permenent middle 14. Cangiano, C. et al. On the stimulation by insulin of 20. Kastin, A. J. & Akerstrom, V. Glucose and insulin
cerebral artery occlusion. J. Cereb. Blood Flow Metab. tryptophan transport across the blood-​brain barrier. increase the transport of leptin through the blood-​
26, 1234–1249 (2006). Biochem. Int. 7, 617–627 (1983). brain barrier in normal mice but not in streptozotocin-​
8. Guo, U. X. et al. 1,25-Dihydroxyvitamin D3 regulates 15. Daniel, P. M., Love, E. R., Moorhouse, S. R. & Pratt, O. E. diabetic mice. Neuroendocrinology 73, 237–242
expression of LRP1 and RAGE in vitro and in vivo, The effect of insulin upon the influx of tryptophan (2001).

NATURe RevIeWS | EndoCrinoLogy volume 15 | AUGUST 2019 | 453


Reviews

21. Ben-​Shachar, D., Yehuda, S., Finberg, J. P., Spanier, I. 45. Dohgu, S., Fleegal-​DeMotta, M. A. & Banks, W. A. 70. Brief, D. J. & Davis, J. D. Reduction of food intake
& Youdim, M. B. Selective alteration in blood-​brain Lipopolysaccharide-​enhanced transcellular transport and body weight by chronic intraventricular insulin
barrier and insulin transport in iron-​deficient rats. of HIV-1 across the blood-​brain barrier is mediated by infusion. Brain Res. Bull. 12, 571–575 (1984).
J. Neurochem. 50, 1434–1437 (1988). luminal microvessel IL-6 and GM-​CSF. J. Neuroinflamm. 71. McGowan, M. K., Andrews, K. M. & Grossman, S. P.
22. May, A. A., Liu, M., Woods, S. C. & Begg, D. P. CCK 8, 167 (2011). Chronic intrahypothalamic infusions of insulin or
increases the transport of insulin into the brain. Physiol. 46. Engstrom, L. et al. Lipopolysaccharide-​induced fever insulin antibodies alter body weight and food intake
Behav. 165, 392–397 (2016). depends on prostaglandin E2 production specifically in the rat. Physiol. Behav. 51, 753–766 (1992).
23. Urayama, A. & Banks, W. A. Starvation and triglycerides in brain endothelial cells. Endocrinology 153, 72. Banks, W. A. & Kastin, A. J. Physiological consequences
reverse the obesity-​induced impairment of insulin 4849–4861 (2012). of the passage of peptides across the blood-​brain
transport at the blood-​brain barrier. Endocrinology 47. Inoue, W. et al. Brain-​specific endothelial induction of barrier. Rev. Neurosci. 4, 365–372 (1993).
149, 3592–3597 (2008). prostaglandin E(2) synthesis enzymes and its temporal 73. Sandoval, D. A., Obici, S. & Seeley, R. J. Targeting the
24. Kastin, A. J., Akerstrom, V. & Maness, L. M. Chronic relation to fever. Neurosci. Res. 44, 51–61 (2002). CNS to treat type 2 diabetes. Nat. Rev. Drug Discov.
loss of ovarian function decreases transport of 48. McCarthy, R. C. & Kosman, D. J. Mechanisms and 8, 386–398 (2009).
leptin into mouse brain. Neurosci. Lett. 310, 69–71 regulation of iron trafficking across the capillary 74. Pocai, A. et al. Hypothalmic K(ATP) channels control
(2001). endothelial cells of the blood-​brain barrier. Front. Mol. hepatic glucose production. Nature 434, 1026–1031
25. Chance, W. T., Balasubramaniam, A., Thomas, I. & Neurosci. 8, 31 (2015). (2005).
Fischer, J. E. Amylin increases transport of tyrosine 49. Miyajima, M. et al. Organic anion transporter 3 75. Biessels, G. J. & Reagan, L. P. Hippocampal insulin
and tryptophan into the brain. Brain Res. 593, 20–24 mediates the efflux transport of an amphipathic organic resistance and cognitive dysfunction. Nat. Rev.
(1992). anion, dehydroepiandrosterone sulfate, across the Neurosci. 16, 660–671 (2015).
26. Banks, W. A. & Kastin, A. J. Modulation of the carrier-​ blood-​brain barrier in mice. Drug Metab. Dispos. 39, 76. Banks, W. A. Insulin in the brain: There and back again.
mediated transport of the Tyr-​MIF-1 across the blood-​ 814–819 (2011). Pharmacol. Ther. 136, 82–93 (2012).
brain barrier by essential amino acids. J. Pharmacol. 50. David, G. F. X. & Kumar, T. C. A. Transfer of steroidal 77. Nelson, T. J., Sun, M. K., Hongpaisan, J. & Alkon, D. L.
Exp. Ther. 239, 668–672 (1986). hormones from blood to the cerebrospinal fluid in the Insulin, PKC signaling pathways and synaptic
27. Urayama, A., Grubb, J. H., Banks, W. A. & Sly, W. S. rhesus monkey. Neuroendocrinology 14, 114–120 remodeling during memory storage and neuronal
Epinephrine enhances lysosomal enzyme delivery (1974). repair. Eur. J. Pharmacol. 585, 76–87 (2008).
across the blood-​brain barrier by up-​regulation of the 51. Marynick, S. P., Haven, W. W., Ebert, M. H. & 78. Biessels, G. J., Bravenboer, B. & Gispen, H. W. Glucose,
mannose 6-phosphate receptor. Proc. Natl Acad. Sci. Loriaux, D. L. Studies on the transfer of steroid insulin and the brain: modulation of cognition and
USA 31, 12873–12878 (2007). hormones across the blood-​cerebrospinal fluid barrier synaptic plasticity in health and disease: a preface.
28. Urayama, A. et al. Alpha adrenergic induction of in Rhesus monkey. Endocrinology 99, 400–405 Eur. J. Pharmacol. 490, 1–4 (2004).
transport of lysosomal enzyme across the blood-​brain (1976). 79. Ferrario, C. R. & Reagan, L. P. Insulin-​mediated synaptic
barrier. PLOS ONE 10, e0142347 (2015). 52. Ohtsuki, S. et al. Dominant expression of androgen plasticity in the CNS: anatomical, functionaland
29. Urayama, A., Grubb, J. H., Sly, W. S. & Banks, W. A. receptors and their functional regulation of organic temporal contexts. Neuropharmacology 136, 182–191
Pharmacologic manipulation of lysosomal enzyme anion transporter 3 in rat brain capillary endothelial (2017).
transport across the blood-​brain barrier. J. Cereb. cells: comparison of gene expression between the 80. Banks, W. A., Kastin, A. J., Huang, W., Jaspan, J. B. &
Blood Flow Metab. 36, 476–476 (2016). blood-​brain and -retinal barriers. J. Cell. Physiol. 204, Maness, L. M. Leptin enters the brain by a saturable
30. Jais, A. et al. Myeloid-​cell-derived VEGF maintains 896–900 (2005). system independent of insulin. Peptides 17, 305–311
brain glucose uptake and limits cognitive impairment 53. Grube, M., Hagen, P. & Jedlitschky, G. Neurosteroid (1996).
in obesity. Cell 165, 882–895 (2016). transport in the brain: role of ABC and SLC transporters. 81. Banks, W. A., Tschop, M., Robinson, S. M. &
31. Fukuda, S. et al. Glucagon-​like peptide-1 strengthens Front. Pharmacol. 9, 354 (2018). Heiman, M. L. Extent and direction of ghrelin transport
the barrier integrity in primary cultures of rat brain 54. Qaiser, M. Z. et al. Uptake and metabolism of sulphated across the blood-​brain barrier is determined by its
endothelial cells under basal and hyperglycemic steroids by the blood-​brain barrier in the adult male rat. unique primary structure. J. Pharmacol. Exp. Ther.
conditions. J. Mol. Neurosci. 29, 211–219 (2016). J. Neurochem. 142, 672–685 (2017). 302, 822–827 (2002).
32. Dohgu, S. et al. Transforming growth factor-·1 55. Bernal, J., Guadano-​Ferraz, A. & Morte, B. Thyroid 82. Diano, S. et al. Ghrelin controls hippocampal spine
upregulates the tight junction and p-​glycoprotein hormone transporters — functions and clinical synapse density and memory performance. Nat.
of brain microvascular endothelial cells. Cell. Mol. implications. Nat. Rev. Endocrinol. 11, 405–416 Neurosci. 9, 381–388 (2006).
Neurobiol. 24, 491–497 (2004). (2015). 83. O’Malley, D. et al. Leptin promotes rapid dynamic
33. Banks, W. A. et al. Triglycerides induce leptin resistance 56. Mayerl, S. et al. Transporters MCT8 and OATP1C1 changes in hippocampal dendritic morphology.
at the blood-​brain barrier. Diabetes 53, 1253–1260 maintain murine brain thyroid hormone homeostasis. Mol. Cell Neurosci. 35, 559–572 (2007).
(2004). J. Clin. Invest. 124, 1987–1999 (2014). 84. Garza, J. C., Guo, M., Zhang, W. & Lu, X. Y. Leptin
34. McCarthy, R. C. & Kosman, D. J. Activation of C6 57. Roberts, L. M. et al. Expression of the thyroid hormone increases adult hippocampal neurogenesis in vivo and
glioblastoma cell ceruloplasmin expression by transporters monocarboxylate transporter-8 (SLC16A2) in vitro. J. Biol. Chem. 283, 18238–18247 (2008).
neighboring human brain endothelia-​derived and organic ion transporter-14 (SLCO1C1) at the blood-​ 85. Greco, S. J. et al. Leptin reduces pathology and
interleukins in an in vitro blood-​brain barrier model brain barrier. Endocrinology 149, 6251–6261 (2008). improves memory in a transgenic mouse model
system. Cell Commun. Signal 12, 65 (2014). 58. Banks, W. A., Kastin, A. J. & Michals, E. A. Transport of Alzheimer’s disease. J. Alzheimers Dis. 19,
35. Yu, C., Kastin, A. J., Tu, H., Waters, S. & Pan, W. of thyroxine across the blood-​brain barrier is directed 1155–1167 (2010).
TNF activates P-​glycoprotein in cerebral microvascular primarily from brain to blood in the mouse. Life Sci. 86. Cuevas, P., Carceller, F., Munoz-​Willery, I. &
endothelial cells. Cell Physiol. Biochem. 20, 853–858 37, 2407–2414 (1985). Gimenez-​Gallego, G. Intravenous fibroblast growth
(2007). 59. Kodding, R., Fuhrmann, H. & van zur Muhlen, A. factor penetrates the blood-​brain barrier and protects
36. Reyes, T. M., Fabry, Z. & Coe, C. L. Brain endothelial Investigations on iodothyronine deiodinase activity in hippocampal neurons against ischemia-​reperfusion
cell production of a neuroprotective cytokine, the maturing brain. Endocrinology 118, 1347–1352 injury. Surg. Neurol. 49, 77–83 (1998).
interleukin-6, in response to noxious stimuli. (1986). 87. Cuevas, P. et al. Central nervous system distribution
Brain Res. 851, 215–220 (1999). 60. Cserr, H. F. & Berman, B. J. Iodide and thiocyanate of fibroblast growth factor injected into the blood
37. Fabry, Z. et al. Production of the cytokines interleukin efflux from brain following injection into rat caudate stream. Neurol. Res. 18, 267–272 (1996).
1 and 6 by murine brain microvessel endothelium nucleus. Am. J. Physiol. 4, F331–F337 (1978). 88. Hsuchou, H., Pan, W. & Kastin, A. J. Fibroblast growth
and smooth muscle pericytes. J. Neuroimmunol. 47, 61. Davson, H. & Hollingsworth, J. R. Active transport of factor 19 entry into brain. Fluids Barriers CNS 10, 32
23–34 (1993). 131 I across the blood-​brain barrier. J. Physiol. 233, (2013).
38. Kis, B. et al. Cerebral endothelial cells are a major 327–347 (1973). 89. Wagner, J. P., Black, I. B. & DiCicco-​Bloom, E.
source of adrenomedullin. J. Neuroendocrinol. 14, 62. Blasberg, R. G. Methotrexate, cytosine arabinoside, and Stimulation of neonatal and adult brain neurogenesis
283–293 (2002). BCNU concentration in brain after ventriculocisternal by subcutaneous injection of basic fibroblast growth
39. Mandi, Y. et al. Nitric oxide production and MDR perfusion. Cancer Treat Rep. 61, 625–631 (1977). factor. J. Neurosci. 19, 6006–6016 (1999).
expression by human brain endothelial cells. 63. Maness, L. M., Banks, W. A., Zadina, J. E. & Kastin, A. J. 90. Bookout, A. L. et al. FGF21 regulates metabolism and
Anticancer Res. 18, 3049–3052 (1998). Periventricular penetration and disappearance of icv circadian behavior by acting on the nervous system.
40. Verma, S., Nakaoke, R., Dohgu, S. & Banks, W. A. Tyr-​MIF-1, DAMGO, tyrosine, and albumin. Peptides 17, Nat. Med. 19, (1147–1152 (2013).
Release of cytokines by brain endothelial cells: 247–250 (1996). 91. Lan, T. et al. FGF19, FGF21, and an FGFR1/β-​klotho-
a polarized response to lipopolysaccharide. Brain 64. Woods, S. C. & Porte, D. Jr. Relationship between activating antibody act on the nervous system to
Behav. Immun. 20, 449–455 (2006). plasma and cerebrospinal fluid insulin levels of dogs. regulate body weight and glycemia. Cell Metab. 26,
41. McGuire, T. R. et al. Release of prostaglandin E-2 in Am. J. Physiol. 233, E331–E334 (1977). 709–718 (2017).
bovine brain endothelial cells after exposure to three 65. Margolis, R. U. & Altszuler, N. Insulin in the 92. Scarlett, J. M. et al. Central injection of fibroblast
unique forms of the antifungal drug amphotericin-​B: cerebrospinal fluid. Nature 215, 1375–1376 (1967). growth factor 1 induces sustained remission of diabetic
role of COX-2 in amphotericin-​B induced fever. Life Sci. 66. Greco, A. V., Ghirlanda, G., Fedeli, G. & Gambassi, G. hyperglycemia in rodents. Nat. Med. 22, 800–806
72, 2581–2590 (2003). Insulin in the cerebro spinal fluid of man. Eur. Neurol. (2016).
42. Ebling, F. J. P. & Lewis, J. E. Tanycytes and hypothalamic 3, 303–307 (1970). 93. Prevot, V. et al. The versatile tanycyte: a hypothalamic
control of energy metabolism. Glia 66, 1176–1184 67. McRory, J. E. & Sherwood, N. M. Ancient divergence integrator of reproduction and energy metabolism.
(2018). of insulin and insulin-​like growth factor. DNA Cell Biol. Endocr. Rev. 39, 333–368 (2018).
43. Ghersi-​Egea, J. F. et al. Molecular anatomy and 16, 939–949 (1997). 94. Johanson, C. E., Duncan, J. A., Stopa, E. G. & Baird, A.
functions of the choroidal blood-​cerebrospinal fluid 68. Bradbury, M. W. B., Segal, M. B. & Wilson, J. Transport Enhanced prospects for drug delivery and brain
barrier in health and disease. Acta Neuropathol. 35, of potassium at the blood-​brain barrier. J. Physiol. targeting by the choroid plexus-​CSF route. Pharm.
337–361 (2018). 221, 617–632 (1972). Res. 22, 1011–1037 (2005).
44. Banks, W. A., Kovac, A. & Morofuji, Y. Neurovascular 69. Maness, L. M., Kastin, A. J., Farrell, C. L. & Banks, W. A. 95. de Lange, E. C. et al. Application of intracerebral
unit crosstalk: pericytes and astrocytes modify cytokine Fate of leptin after intracerebroventricular injection microdialysis to study regional distribution kinetics of
secretion patterns of brain endothelial cells. J. Cereb. into the mouse brain. Endocrinology 139, 4556–4562 drug in rat brain. Br. J. Pharmacol. 116, 2538–2544
Blood Flow Metab. 38, 1104–1118 (2018). (1998). (1995).

454 | AUGUST 2019 | volume 15 www.nature.com/nrendo


Reviews
96. Zlokovic, B. V. et al. Differential regulation of leptin 117. Schulz, G. B. et al. Cerebral cavernous malformation-1 141. Rutenberg, G. W. et al. Body composition in baboons:
transport by the choroid plexus and blood-​brain protein controls DLL4-Notch3 signalling between the evaluating a morphometric method. Am. J. Primatol.
barrier and high affinity transport systems for entry endothelium and pericytes. Stroke 46, 1337–1343 12, 275–285 (1987).
into hypothalamus and across the blood-​cerebrospinal (2015). 142. Bribiescas, R. G. Serum leptin levels in Ache Amerindian
fluid barrier. Endocrinology 141, 1434–1441 (2000). 118. Kaiyala, K. J., Prigeon, R. L., Kahn, S. E., Woods, S. C. females with normal adiposity are not significantly
97. Nonaka, N., Hileman, S. M., Shioda, S., Vo, P. & & Schwartz, M. W. Obesity induced by a high-​fat diet different from American anorexia nervosa patients.
Banks, W. A. Effects of lipopolysaccharide on leptin is associated with reduced brain insulin transport in Am. J. Hum. Biol. 17, 207–210 (2005).
transport across the blood-​brain barrier. Brain Res. dogs. Diabetes 49, 1525–1533 (2000). 143. Kirchengast, S. Weight status of adult !Kung San and
1016, 58–65 (2004). 119. Mooradian, A. D. Blood-​brain barrier choline transport Kavango people from northern Namibia. Ann. Hum.
98. Banks, W. A. & Farrell, C. L. Impaired transport of is reduced in diabetic rats. Diabetes 36, 1094–1097 Biol. 25, 541–551 (1996).
leptin across the blood-​brain barrier in obesity is (1987). 144. Schwartz, M. W. et al. Cerebrospinal fluid leptin levels:
acquired and reversible. Am. J. Physiol. 285, E10–E15 120. Hong, H. et al. Downregulation of LRP1 at the blood-​ relationship to plasma levels and adiposity in humans.
(2003). brain barrier in streptozotocin-​induced diabetic mice. Nat. Med. 2, 589–593 (1996).
99. Banks, W. A. et al. Triglycerides cross the blood-​brain Neuropharmacology 56, 1054–1059 (2009). 145. Caro, J. F. et al. Decreased cerebrospinal-​fluid/serum
barrier and induce central leptin and insulin receptor 121. Banks, W. A., DiPalma, C. R. & Farrell, C. L. Impaired leptin ratio in obesity: a possible mechanism for leptin
resistance. Int. J. Obes. (Lond.) 42, 391–397 (2018). transport of leptin across the blood-​brain barrier in resistance. Lancet 348, 159–161 (1996).
100. Romero, L. I., Kakucska, I., Lechan, R. M. & Reichlin, S. obesity. Peptides 20, 1341–1345 (1999). 146. Mantzoros, C., Flier, J. S., Lesem, M. D., Brewerton, T. D.
Interleukin-6 (IL-6) is secreted from the brain after 122. Ouyang, S. et al. Diet-​induced obesity suppresses & Jimerson, D. C. Cerebrospinal fluid leptin in anorexia
intracerebroventricular injection of IL-1β in rats. Am. J. expression of many proteins at the blood-​brain barrier. nervosa: Correlation with nutritional status and potential
Physiol. 270, R518–R524 (1996). J. Cereb. Blood Flow Metab. 34, 43–51 (2014). role in resistance to weight gain. J. Clin. Endocrinol.
101. Chen, G. & Reichlin, S. Clearance of [125 I]-tumor 123. Mooradian, A. D. & Smith, T. L. The effect of Metab. 82, 1845–1851 (1997).
necrosis factor-​alpha from the brain into the blood experimentally induced diabetes mellitus on the lipid 147. Smith, Q. R., Momma, S., Aoyagi, M. & Rapoport, S. I.
after intracerebroventricular injection into rats. order and composition of rat cerebral microvessels. Kinetics of neutral amino acid transport across the
Neuroimmunomodulation 5, 261–269 (1998). Neurosci. Lett. 145, 145–148 (1992). blood-​brain barrier. J. Neurochem. 49, 1651–1658
102. Chen, G., McCuskey, R. S. & Reichlin, S. Blood 124. Beard, R. S. Jr, Reynolds, J. J. & Bearden, S. E. (1987).
interleukin-6 and tumor necrosis factor-​alpha Hyperhomocysteinemia increases permeability of the 148. Fernstrom, J. D. Branched-​chain amino acids and
elevation after intracerebroventricular injection of blood-​brain barrier by NMDA receptor-​dependent brain function. J. Nutr. 135, 1439S–1546S (2005).
Escherichia coli endotoxin in the rat is determined regulation of adherens and tight junctions. Blood 118, 149. van Spronsen, F. J., de Groot, M. J., Hoeksma, M.,
by two opposing factors: peripheral induction by LPS 2007–2014 (2011). Reijngoud, D.-J. & van Rijn, M. Large neutral amino
transferred from brain to blood and inhibition of 125. Rhodehouse, B. C., Mayo, J. N., Beard, R. S. Jr, acids in the treatment of PKU: from theory to practice.
peripheral response by a brain-​mediated mechanism. Chen, C. H. & Bearden, S. E. Opening of the blood-​ J. Inherit. Metab. Dis. 33, 671–676 (2010).
Neuroimmunomodulation 8, 59–69 (2000). brain barrier before cerebral pathology in mild 150. Banks, W. A., Burney, B. O. & Robinson, S. M. Effects
103. Logsdon, A. F., Erickson, M. A., Rhea, E. M., hyperhomocysteinemia. PLOS ONE 8, e63951 of triglycerides, obesity, and starvation on ghrelin
Salameh, T. S. & Banks, W. A. Gut reactions: how the (2013). transport across the blood-​brain barrier. Peptides 29,
blood-​brain barrier connects the microbiome and 126. Delange, F. The disorders induced by iodine deficiency. 2061–2065 (2008).
the brain. Exp. Biol. Med. (Maywood) 243, 159–165 Thyroid 4, 107–128 (1994). 151. Holscher, C. Novel dual GLP-1/GIP receptor agonists
(2018). 127. Nunez, B. et al. Cerebral cortex hyperthyroidism of show neuroprotective effects in Alzheimer’s and
104. Erickson, M. A. & Banks, W. A. Neuroimmune axes newborn mct8-deficient mice transiently suppressed Parkinson’s disease models. Neuropharmacology
of the blood-​brain barriers and blood-​brain interfaces: by lat2 inactivation. PLOS ONE 9, e96915 (2014). 136, 251–259 (2018).
bases for physiological regulation, disease states, 128. Heni, M., Kullmann, S., Preissl, H., Fritsche, A. & 152. Gejl, M. et al. Blood-​brain glucose transfer in
and pharmacological interventions. Pharmacol. Rev. Haring, H. U. Impaired insulin action in the human Alzheimer’s disease: effect of GLP-1 analog treatment.
70, 278–314 (2018). brain: causes and metabolic consequences. Nat. Rev. Sci. Rep. 7, 17490 (2017).
105. Sampson, T. R. et al. Gut microbioata regulate Endocrinol. 11, 701–711 (2015). 153. Tamargo, I. A. et al. Novel GLP-1R/GIPR co-​agonist
motor deficits and neuroinflammation in a model of 129. De Vivo, D. C. et al. Defective glucose transport “twincretin” is neuroprotective in cell and rodent
Parkinson’s disease. Cell 167, 1469–1480 (2016). across the blood-​brain barrier as a cause of persistent models of mild traumatic brain injury. Exp. Neurol.
106. Starr, J. M. et al. Increased blood-​brain barrier hypoglycorrhachia, seizures, and developmental delay. 288, 176–186 (2017).
permeability in type II diabetes demonstrated by N. Engl. J. Med. 325, 703–709 (1991). 154. Deli, M. A., Abraham, C. R., Kataoka, Y. & Niwa, M.
gadolinium magnetic resonance imaging. J. Neurol. 130. Craft, S. et al. Cerebrosinal fluid and plasma insulin Permeability studies on in vitro blood-​brain barrier
Neurosurg. Psychiatry 74, 70–76 (2003). levels in Alzheimer’s disease: relationship to severity of models: physiology, pathology, and pharmacology.
107. Huber, J. D., VanGilder, R. L. & Houser, K. A. dementia and apolipoprotein E genotype. Neurology Cell. Mol. Neurobiol. 25, 59–127 (2005).
Streptozotocin-​induced diabetes progressively increases 50, 164–168 (1998). 155. Wolburg, H., Noell, S., Mack, A., Wolburg-​Buchholz, K.
blood-​brain barrier permeability in specific brain 131. Talbot, K. et al. Demonstrated brain insulin resistance & Dallier-​Becker, P. Brain endothelial cells and the glio-
regions in rats. Am. J. Physiol. 291, H2660–H2668 in Alzheimer’s disease patients is associated with ​vascular complex. Cell Tissue Res. 335, 75–96 (2009).
(2006). IGF-1 resistance, IRS-1 dyregulation, and cognitive 156. Saunders, N. R., Dziegielewska, K. M., Mollgard, K.
108. Abuhaiba, S. et al. Occipital blood-​brain barrier decline. J. Clin. Invest. 122, 1316–1338 (2012). & Habgood, M. D. Physiology and molecular biology
permeability is an independent predictor of visual 132. Sartorius, T. et al. The brain response to peripheral of barrier mechanisms in the fetal and neonatal brain.
outcome in type 2 diabetes, irrespective of the retinal insulin declines with age: a contribution of the blood-​ J. Physiol. 596, 5723–5756 (2018).
barrier: a logitudinal study. J. Neuroendocrinol. 30, brain barrier? PLOS ONE 10, e0126804 (2015). 157. Cornford, E. M., Braun, L. D., Oldendorf, W. H.
e12566 (2018). 133. Pelleymounter, M. A. et al. Effects of the obese gene & Hill, M. A. Comparison of lipid-​mediated blood-​
109. Huber, J. D. Diabetes, cognitive function, and the product on body weight regulation in ob/ob mice. brain-barrier penetrability in neonates and adults.
blood-​brain barrier. Curr. Pharm. Des. 14, 1594–1600 Science 269, 540–543 (1995). Am. J. Physiol. 243, C161–C168 (1982).
(2008). 134. Banks, W. A., Clever, C. M. & Farrell, C. L. Partial 158. Dickson, P. W., Aldred, A. R., Marley, P. D., Bannister, D.
110. Takechi, R. et al. Blood-​brain barrier dysfunction saturation and regional variation in the blood to & Schreiber, G. Rat choroid plexus specializes in
precedes cognitive decline and neurodegeneration in brain transport of leptin in normal weight mice. the synthesis and the secretion of transthyretin
diabetic insulin resistant mouse model: an implication Am. J. Physiol. 278, E1158–E1165 (2000). (prealbumin). Regulation of tranthyretin synthesis in
for causal link. Front. Aging Neurosci. 9, 399 (2017). 135. Licinio, J. et al. Phenotypic effects of leptin replacement choroid plexus is indpendent from that in liver. J. Biol.
111. Hammes, H. P. et al. Pericytes and the pathogenesis on morbid obesity, diabetes mellitus, hypogonadism, Chem. 15, 3475–3478 (1986).
of diabetic retinopathy. Diabetes 51, 3107–3112 and behavior in leptin-​deficient adults. Proc. Natl Acad. 159. Zhao, F. et al. Effects of passage and cryopreservation
(2002). Sci. USA 101, 4531–4536 (2004). on neurotophic factor secretion from choroid plexus
112. Price, T. O., Eranki, V., Banks, W. A., Ercal, N. & 136. Ingalls, A. M., Dickie, M. M. & Snell, G. D. Obese, epithelial cells. Biomed. Rep. 8, 535–539 (2018).
Shah, G. N. Topiramate treatment protects blood-​ a new mutation in the house mouse. J. Hered. 41, 160. Broadwell, R. D. & Sofroniew, M. V. Serum proteins
brain barrier pericytes from hyperglycemia-​induced 317–318 (1950). bypass the blood-​brain barrier for extracellular entry
oxidative damage in diabetic mice. Endocrinology 137. Bribiescas, R. G. Serum leptin levels and anthropometric to the central nervous system. Exp. Neurol. 120,
153, 362–372 (2012). correlates in Ache Amerindians of Eastern Paraguay. 245–263 (1993).
113. Dore-​Duffy, P. Pericytes: pluripotent cells of the blood Am. J. Phys. Anthropol. 115, 297–303 (2001).
brain barrier. Curr. Pharm. Des. 14, 1581–1593 138. Halaas, J. L. et al. Physiological response to long-​term Competing interests
(2008). peripheral and central leptin infusion in lean and obese The author declares no competing interests.
114. Daneman, R., Zhou, L., Kebede, A. A. & Barres, B. A. mice. Proc. Natl Acad. Sci. USA 94, 8878–8883
Pericytes are required for blood-​brain barrier integrity (1997). Publisher’s note
during embryogenesis. Nature 468, 562–566 (2010). 139. van Heek, M. et al. Diet-​induced obese mice develop Springer Nature remains neutral with regard to jurisdictional
115. Zhang, Y. et al. Involvement of PUMA in pericyte peripheral, but not central, resistance to leptin. J. Clin. claims in published maps and institutional affiliations.
migration induced by methamphetamine. Exp. Cell Invest. 99, 385–390 (1997).
Res. 356, 28–39 (2017). 140. El Haschimi, K., Pierroz, D. D., Hileman, S. M., Reviewer information
116. Sagare, A. P. et al. Pericyte loss influences Alzheimer-​ Bjorbaek, C. & Flier, J. S. Two defects contribute to Nature Reviews Endocrinology thanks B. Levin, D. Begley
like neurodegeneration in mice. Nat. Commun. 4, hypothalamic leptin resistance in mice with diet-​induced and V. Prevot, and the other anonymous reviewers, for their
2932 (2013). obestiy. J. Clin. Invest. 105, 1827–1832 (2000). contribution to the peer review of this work.

NATURe RevIeWS | EndoCrinoLogy volume 15 | AUGUST 2019 | 455

You might also like