William A. Bankscorresponding author

Endocrinology. 2012 Sep; 153(9): 4111–4119.Published online 2012 Jul 9.

Abstract

The blood-brain barrier (BBB) separates the central nervous system (CNS) from the peripheral tissues. However, this does not prevent hormones from entering the brain, but shifts the main control of entry to the BBB. In general, steroid hormones cross the BBB by transmembrane diffusion, a nonsaturable process resulting in brain levels that reflect blood levels, whereas thyroid hormones and many peptides and regulatory proteins cross using transporters, a saturable process resulting in brain levels that reflect blood levels and transporter characteristics. Protein binding, brain-to-blood transport, and pharmacokinetics modulate BBB penetration. Some hormones have the opposite effect within the CNS than they do in the periphery, suggesting that these hormones cross the BBB to act as their own counterregulators. The cells making up the BBB are also endocrine like, both responding to circulating substances and secreting substances into the circulation and CNS. By dividing a hormone's receptors into central and peripheral pools, the former of which may not be part of the hormone's negative feed back loop, the BBB fosters the development of variable hormone resistance syndromes, as exemplified by evidence that altered insulin action in the CNS can contribute to Alzheimer's disease. In summary, the BBB acts as a regulatory interface in an endocrine-like, humoral-based communication between the CNS and peripheral tissues.

The brain binds and secretes circulating substances (1, 2). As such, it qualifies as an endocrine organ, being one of the largest and the most metabolically active of the endocrine organs, acting as both a target and secretor of hormones. One of many unique aspects of the endocrine brain is that its capillary bed is modified to prevent leakage, thus preventing the production of an ultrafiltrate (3). This modified capillary bed comprises the vascular blood-brain barrier (BBB), and the loss of ultrafiltrate production precludes the main mechanism by which hormones gain access to the interstitial fluid of their target tissues.

However, the BBB does not negate the influence of hormones on the central nervous system (CNS). Instead, the BBB redefines how hormones access the CNS and, in so doing, the BBB becomes an important conduit and interface in humoral-based brain-body communication. This review will examine the mechanisms used by hormones to cross the BBB, some of the resulting physiological and pathological consequences of these mechanisms, and some disease states that can arise when brain-body communication at the level of the BBB goes awry.

Mechanisms of Hormonal Translocation Across the BBB

Nearly every mechanism by which a substance can cross or interact with the BBB is used by one hormone or the other (Fig. 1). In general, steroid hormones cross the BBB by transmembrane diffusion whereas thyroid hormones, peptide hormones, and regulatory proteins cross by saturable systems. Their small size and lipid solubility allow steroid hormones to easily cross the BBB bidirectionally (i.e. in the brain-to-blood and blood-to-brain directions) by the process of transmembrane diffusion (4). Steroid hormones enter rapidly and throughout the brain by this nonsaturable process, the bidirectional and nonsaturable nature of entry allowing brain levels to come into rapid equilibrium with and to directly mirror blood levels, the level of brain retention being dependent more on the presence of receptors than on BBB permeability. Peptides can also cross to some degree by transmembrane diffusion, and enzymatically stable analogs can exert effects on the CNS (59).

The rate at which hormones transported by saturable mechanisms cross the BBB vary greatly as a function of the characteristics and distribution of the transporter. In general, the transport rate for the primary ligand of a transporter crosses about 10–40 times faster than it would were it to depend on transmembrane diffusion (10). Likewise, regional hormonal uptake and retention are influenced by the anatomical distribution of the transporter and its avidity for its ligand. The saturable aspect means that brain levels are not necessarily a linear reflection of blood levels both because of the nonlinear nature of transporters and because transporter rates can be modulated by many factors. Transporter dysfunction can lead to CNS dysfunction as discussed below. The list of regulatory substances that cross the BBB by way of saturable transporters is long and growing (1115). Although peptide and regulatory protein hormones were once assumed to not penetrate the BBB, it is now clear that they do so by both saturable and nonsaturable mechanisms (16). For example, saturable transport systems have been described for thyroid hormones, peptide and protein feeding hormones, cytokines, and chemokines (13, 17, 18).

Transporters can also remove substances from the brain (19). In some cases, these transporters prevent blood-borne substances from entering the brain and in other cases they transport substances of CNS origin into the blood stream. In mice, T4 is primarily transported in the brain-to-blood direction with T3 being the primary thyroid hormone transported into the brain (20). This differential transport of thyroid hormones has been suggested to explain in part why T3 may be superior to T4 in the treatment of critically ill hypothyroid patients (21). P-glycoprotein is an important efflux pump for many xenobiotics (22), but its ligands include some steroid hormones such as testosterone and progesterone, but not others such as cortisol and β-estradiol (23, 24).

The brain-to-blood movement of biologically active substances means that the CNS can act as an endocrine-secreting organ. Both IL-6 and TNF-α of CNS origin can contribute significantly to the levels in blood (2528). Corticotrophin releasing factor transported into blood from brain can affect β-endorphin production by the spleen (29). Other substances transported into the blood from brain that likely contribute significantly to blood levels include IL-2, amyloid β peptide, endorphins, and enkephalins (18, 3033).

Some very large hormones may cross the BBB by the residual leakiness of the BBB, termed the extracellular pathways (34). The sites for this leakage include the pial surface, the Virchow-Robin spaces, and the subpial cortical gray matter. Erythropoietin, antibodies, binding proteins, and albumin are examples of substances that cross by this mechanism (35). The pharmacokinetics favorable for this pathway have long half-lives in blood and small volumes of distribution. Passage is nonsaturable but independent of lipid solubility or molecular size, and the rate of penetration is low.

Effects of Binding Proteins on Hormone Penetration of the BBB

Albumin and binding proteins influence hormonal penetration across the BBB (36, 37). Cerebrospinal fluid (CSF) levels of albumin are about 0.5% of those in serum, yet this is enough to produce free and bound fractions of hormones in the CSF and brain interstitial fluid. For example, δ-sleep-inducing peptide is about 90% bound in blood and about 50% bound in CSF (38). Binding proteins are very influential in the passage of substances that cross the BBB by way of transmembrane diffusion with only free or loosely-bound hormone being available for transport. For substances with transporters, the relative avidity of the binding protein vs. the transporter dictates the degree to which the bound pool is available for passage across the BBB.

A dramatic example of the interplay between transporter function and binding proteins is that of T4, a hormone that is transported by a saturable mechanism into the CSF. The CSF/serum ratio of total T4 in dogs is 0.025, but the ratio for free T4 is 2.4 (17). In this case, the major source of CSF transthyretin, an important thyroid-binding protein, is not the circulation but the choroid plexus, the anatomical seat of the blood-CSF barrier, with the choroid plexus synthesizing and secreting transthyretin into the CSF (39).

Binding hormones alter pharmacokinetic aspects of their ligands, such as circulating half-life, that ultimately influence the rate and degree of penetration. Sex-binding globulin, for example, slows the rate at which testosterone crosses the BBB but also decreases clearance from blood, allowing more time for testosterone to penetrate the BBB and enter the CNS, ultimately having little affect on the total amount of testosterone entering the brain. However, lower, sustained levels of receptor occupancy vs. short-term, higher receptor occupancy can greatly influence hormone action, producing very different pharmacodynamic profiles (40).

These effects of binding hormones affect the pharmacokinetic and pharmacodynamic profiles of drugs derived from the parent hormones. For example, protein binding of insulin detemir prolongs its half-life in blood and its hypoglycemic effects, while decreasing its effects on body weight gain and altering its CNS effects (4144). The protein binding, however, prevents insulin detemir from rapidly crossing the BBB (45).

Modulation of Saturable Transporter Mechanisms and Targeting to Brain Regions

The characteristics of the saturable transport of hormones are markedly different from those of metabolic substances. Representative of metabolic substances, glucose is needed by every region of the brain and in large amounts, and a high rate of transport throughout the CNS reflects this need. In comparison, the transport rate for a hormone can vary greatly among brain regions. As examples: some regions of the brain do not have measurable rates of insulin transport (46); the transport rate for TNF-α varies among brain regions by 10-fold (47); transport of leptin occurs throughout the CNS but with a high concentration at the arcuate nucleus (48); IL-1 α has a concentrated uptake at the posterior division of the septum (49) and marked differences in transport into brain vs. spinal cord (13). In essence then, the transport of regulatory substances is much more variable, assumedly reflecting a targeting to areas of hormone activity. For example, the high rate of uptake of IL-1 into the posterior division of the septum coordinates with its ability to modulate memory (50).

Many of the hormonal transporters are not static but are themselves regulated. This regulation is presumed to reflect responses of the BBB to the needs of the CNS and therefore are, on one level, physiological. In some cases, this regulation contributes to premorbid conditions and disease states (Table 1). For example, impaired transport of leptin in obesity contributes to the leptin resistance seen in that condition (5153). Triglycerides, insulin, glucose, estrogen, and epinephrine are some of the modulators of leptin transport that likely contribute to the regulation of the leptin transporter (5457). Lower CSF and CSF/serum ratios of insulin suggest a decrease in insulin transport in pregnancy and could underlie the insulin-remediable defect in baroreceptor response seen in pregnancy (58, 59).

The BBB as an Endocrine Tissue

The cells that form the BBB both respond to hormones in the circulation and also secrete substances into the circulation. Examples of the BBB responding to circulating hormones are numerous. Insulin affects several parameters of brain endothelial cell function including modulation of amino acid, leptin, and the p-glycoprotein transporters (56, 6062) and affects alkaline phosphatase and glutamate cysteine ligase catalytic subunit, the latter important in maintaining intracellular levels of glutathione (63, 64). Estradiol alters the Na-K-Cl cotransporter at the BBB (65). Angiotensin II affects vesicular trafficking of brain endothelial cells (66). Circulating epinephrine enhances leptin and enzyme transport across the BBB (67, 68). TNF-α and endothelin modulate the brain-to-blood efflux transporter p-glycoprotein (69, 70).

The study of the BBB as secretor is less well explored. However, brain endothelial cells secrete cytokines, chemokines, nitric oxide, adrenomedullin, and prostaglandins both constitutively and in response to treatment with lipopolysaccharide, cytokines, and other substances (7180).

A unique feature of the BBB that relies on it having cell membrane surfaces facing both into the blood stream and into the interstitial fluid of the CNS is that it can receive signals from one compartment and secret regulatory substances into the other. For example, lipopolysaccharide applied to the brain (abluminal) side of the brain endothelial cell (BEC) stimulates them to increase their release of IL-6 from the blood (luminal) side (81). Adiponectin applied to the luminal membrane of BECs inhibits IL-6 secretion from the abluminal membrane, providing a mechanism by which the adipogenic effects of adiponectin could be relayed into the brain (82).

These endocrine effects at the BBB likely also contribute to disease states. The effects of angiotensin II on vesicular trafficking across brain endothelial cells (66) may underpin BBB disruption in hypertensive encephalopathy (83) as mediated by the renin-angiotensin-aldosterone system (84). IL-1 induces the fever seen in sickness behavior by inducing the secretion of prostaglandins from brain endothelial cells (79, 8587). Up-regulation of adrenergic receptors at BBB cells in Alzheimer's patients could be a basis for, or in response to, the cerebrovascular abnormalities seen in that disease (88). Through its regulation of Na-K-Cl channels, estradiol reduces brain edema after middle cerebral artery occlusion, providing a mechanism that may underlie the effects of estrogens on stroke and stroke outcomes (65).

Unique Interactions: Counterregulatory and Potentiating Effects

Hormones transported across the BBB can act within the CNS to modulate their peripheral effects (Fig. 2). As appreciated for almost two decades, many peptides that are transported across the BBB have effects in the brain opposite to those produced at peripheral tissues (89). For example, CNS insulin has been associated with an increase in serum glucose and a decrease in feeding (9093); administration of cholecystokinin-8 into the CNS is associated with an attenuation of passive avoidance behavior whereas peripheral administration is associated with facilitation (94); CNS calcitonin enhances whereas peripheral calcitonin inhibits glucose-stimulated insulin release (95); and CNS TRH simulates whereas peripheral TRH decreases locomotor activity (9698). This suggests that these hormones may be acting as their own counterregulatory hormones, the main effect being induced at peripheral tissues and a secondary, counterregulatory effect being induced within the CNS by the small amount of hormone that crosses the BBB.

Other hormones have effects in the CNS that are consistent with their peripheral effects. Secretin was the first hormone to be discovered, stimulating pancreatic secretion when administered peripherally (99). Secretin also affects CNS function either when given peripherally (100) or directly into brain (101). Secretin is found in the brain (102, 103) in part because of CNS synthesis (104) and in part because it is transported into the CNS, possibly across the choroid plexus rather than at the vascular barrier (105), where it acts at its CNS receptors (106, 107), stimulating cAMP and facilitating γ-aminobutyric acid neurotransmission (108, 109). Interestingly, CNS secretin also stimulates pancreatic secretion, but at much lower doses than those required when administered peripherally (110). About half of the effect that systemic insulin exerts on hepatic glucose production may be derived from insulin crossing the BBB and activating the vagus through ATP-sensitive potassium channels in the hypothalamus (111). Some hormones cross the BBB to stimulate their own secretion from CNS sources. For example, TNF-α transported across the BBB stimulates release of TNF from CNS sources, increasing to levels that can be toxic to dopaminergic cells of the substantia nigra, thus promoting the development of Parkinson's disease (112). In contrast to the paradoxic effect outlined above, this suggests that other hormones cross the BBB to reinforce or potentiate their peripheral effects through CNS mechanisms.

Unique Interactions: The Question of Variable Hormone Resistance

Hormone resistance syndromes have been appreciated since the description of pseudohypoparathyroidism (113, 114). The variable hormone resistance syndromes for thyroid hormones illustrate that the degree of resistance may not be the same at all of the sites of action for a hormone (115). The presence of a BBB, which first divides the body into central and peripheral compartments and then defines and regulates the interactions between those compartments, fosters the development of hormonal resistance arising at three sites: 1) the CNS compartment, 2) the peripheral tissue compartment, and 3) the BBB. The relative resistances of tissues within and among these compartments may play a role in various diseases, including obesity and Alzheimer's disease.

In humans and animals who do not make leptin, its administration causes profound weight loss (116, 117). However, in humans and in animal models of diet-induced obesity, resistance to leptin limits its usefulness as an anorectic (118). Resistance to leptin's anorectic effects arises at two levels: at the BBB and at brain receptors (119). Modeling based on CSF and serum levels of leptin suggests that, although resistance at both of these sites is progressive with increasing obesity, early in obesity the dominant site of resistance is at the BBB (120). Studies of the progressive resistance to leptin given by the CNS and peripheral routes to animals with diet-induced obesity further support this (52, 53).

Diabetes mellitus type III is a term that emphasizes that Alzheimer's disease has been associated with a defect in insulin action within the CNS, either because of decreased levels of insulin in the brain or because of resistance at CNS insulin receptors (121125). Several studies have shown that delivery of insulin to brain by way of peripheral or intranasal administration can improve cognition in normal subjects, persons with memory impairment, and in patients with Alzheimer's disease (126130). Although diabetes mellitus type 2 is a risk factor for Alzheimer's disease, there are many patients with diabetes type 2 who do not have Alzheimer's disease and many patients with Alzheimer's disease who do not have diabetes. This would suggest that to the degree that Alzheimer's disease relates to CNS insulin deficiency, that deficiency can be independent of peripheral insulin resistance.

A mismatch between the degree of CNS and peripheral resistances to insulin would illustrate an interesting dilemma in hormonal regulation. In the case of partial peripheral insulin resistance, the pancreas can compensate by increasing insulin secretion until glucose returns to a normal range. But CNS receptors, especially those not related to glucose sensing, are not involved in the negative feedback loop that exists in the periphery between glucose and insulin. Furthermore, at some point, elevations in serum insulin are not translated into equivalent elevations in CNS insulin because the insulin transporter at the BBB becomes saturated. As a result of these two phenomena, how or even if the CNS can compensate for an isolated partial resistance to insulin is unclear.

Conclusions

The BBB's unique location that places it simultaneously in the peripheral circulation and in the CNS and its unique functions of first separating the CNS from the periphery and then regulating their humoral interactions gives the BBB unusual endocrine properties. The cells that compose the BBB themselves also qualify as endocrine tissues, both responding to and secreting circulating substances. Endocrine conditions and diseases such as diabetes mellitus and obesity lead to BBB dysfunction, and dysfunctions of the endocrine BBB contribute to these states. These characteristics allow for unique interactions among the BBB, peripheral tissues, and CNS.

Acknowledgments

We thank Emily Wing (SIBCR and GRECC, Veterans Affairs Puget Sound Health Care System) for help with the graphics.

This work was supported by National Institutes of Health grants RO1 DK083485, R01 AG029839, R01 NS051134, and Veterans Affairs Merit Review.

Disclosure Summary: There are no conflicts of interest regarding this manuscript.

Footnotes

Abbreviations:

BBB
Blood-brain barrier
BEC
brain endothelial cell
CNS
central nervous system
CSF
cerebrospinal fluid.


References


1. Kastin AJ, Pan W, Zadina JE, Banks WA. 2001. The endocrine brain. In: Becker KL, editor. , ed. Principles and practice of endocrinology and metabolism. Philadelphia: Lippincott Williams & Wilkins; 1611–1615
2. Morley JE, Silver AJ. 1991. Role of the endocrine brain in the control of eating and drinking. In: Motta M, editor. , ed. Brain endocrinology. 2nd ed New York: Raven Press; 431–447
3. Davson H, Segal MB. 1996. Physiology of the CSF and blood-brain barriers. Boca Raton, FL: CRC Press
4. Cornford EM, Braun LD, Oldendorf WH, Hill MA. 1982. Comparison of lipid-mediated blood-brain-barrier penetrability in neonates and adults. Am J Physiol243:C161–C168
5. Banks WA, Kastin AJ. 1985. Peptides and the blood-brain barrier: lipophilicity as a predictor of permeability. Brain Res Bull 15:287–292
6. Banks WA, Kastin AJ, Coy DH. 1984. Evidence that [125 I]N-Tyr-δ sleep-inducing peptide crosses the blood-brain barrier by a non-competitive mechanism. Brain Res301:201–207
7. Gray RA, Vander Velde DG, Burke CJ, Manning MC, Middaugh CR, Borchardt RT. 1994. Delta-sleep-inducing peptide: solution conformational studies of a membrane-permeable peptide. Biochemistry 33:1323–1331
8. Miller LH, Turnbull BA, Kastin AJ, Coy DH. 1986. Sleep-wave activity of a δ sleep-inducing peptide analog correlates with its penetrance of the blood-brain barrier. Sleep 9:80–84
9. Kastin AJ, Pearson MA, Banks WA. 1991. EEG evidence that morphine and an enkephalin analog cross the blood-brain barrier. Pharmacol Biochem Behav 40:771–774
10. Oldendorf WH. 1971. Brain uptake of radio-labelled amino acids, amines and hexoses after arterial injection. Am J Physiol 221:1629–1639
11. Kastin AJ, Pan W. 2003. Feeding peptides interact in several ways with the blood-brain barrier. Curr Pharm Des 9:789–794
12. Pan W, Kastin AJ. 2004. Transport of cytokines and neurotrophins across the blood-brain barrier and their regulation after spinal cord injury. In: Sharma HS, Westman J, editors. , eds. Blood-spinal cord and brain barriers in health and disease. Amsterdam: Elsevier; 395–407
13. Pan W, Kastin AJ. 2008. Cytokine transport across the injured blood-spinal cord barrier. Curr Pharm Des 14:1620–1624
14. Davson H, Segal MB. 1996. Special aspects of the blood-brain barrier. In: Physiology of the CSF and blood-brain barriers. Boca Raton, FL: CRC Press; 303–485
15. Brownlees J, Williams CH. 1993. Peptidases, peptides, and the mammalian blood-brain barrier. J Neurochem 60:793–803
16. Banks WA, Kastin AJ. 1996. Passage of peptides across the blood-brain barrier: pathophysiological perspectives. Life Sci 59:1923–1943
17. Hagen GA, Elliot TM. 1973. Transport of thyroid hormones in serum and cerebrospinal fluid. J Clin Endocrinol Metab 37:415–422
18. Banks WA, Kastin AJ. 1990. Editorial Review: Peptide transport systems for opiates across the blood-brain barrier. Am J Physiol 259:E1–E10
19. Taylor EM. 2002. The impact of efflux transporters in the brain on the development of drugs for CNS disorders. Clin Pharmacokinet 41:81–92
20. Banks WA, Kastin AJ, Michals EA. 1985. Transport of thyroxine across the blood-brain barrier is directed primarily from brain to blood in the mouse. Life Sci37:2407–2414
21. Chernow B, Burman KD, Johnson DL, McGuire RA, O'Brian JT, Wartofsky L, Georges LP. 1983. T 3 may be a better agent than T 4 in the critically ill hypothyroid patient: evaluation of transport across the blood-brain barrier in a primate model. Crit Care Med 11:99–104
22. Begley DJ. 2004. ABC transporters and the blood-brain barrier. Curr Pharm Des 10:1295–1312
23. Tsuji A, Terasaki T, Takabatake Y, Tenda Y, Tamai I, Yamashima T, Moritani S, Tsuruo T, Yamashita J. 1992. P-glycoprotein as the drug efflux pump in primary cultured bovine brain capillary endothelial cells. Life Sci 51:1427–1437
24. Mason BL, Pariante CM, Jamel S, Thomas SA. 2010. Central nervous system (CNS) delivery of glucocorticoids is fine-tuned by saturable transporters at the blood-CNS barriers and nonbarrier regions. Endocrinology 151:5294–5305
25. Chen G, Castro WL, Chow HH, Reichlin S. 1997. Clearance of 125 I-labelled interleukin-6 from brain into blood following intracerebroventricular injection in rats. Endocrinology 138:4830–4836
26. Chen G, McCuskey RS, Reichlin S. 2000. Blood interleukin-6 and tumor necrosis factor-α elevation after intracerebroventricular injection of Escherichia coli endotoxin in the rat is determined by two opposing factors: peripheral induction by LPS transferred from brain to blood and inhibition of peripheral response by a brain-mediated mechanism. Neuroimmunomodulation 8:59–69
27. Chen G, Reichlin S. 1998. Clearance of [125 I]-tumor necrosis factor-α from the brain into the blood after intracerebroventricular injection into rats. Neuroimmunomodulation 5:261–269
28. Romero LI, Kakucska I, Lechan RM, Reichlin S. 1996. Interleukin-6 (IL-6) is secreted from the brain after intracerebroventricular injection of IL-1· in rats. Am J Physiol 270:R518–R524
29. Martins JM, Banks WA, Kastin AJ. 1997. Transport of CRH from mouse brain directly affects peripheral production of β-endorphin by the spleen. Am J Physiol273:E1083–E1089
30. Banks WA, Niehoff ML, Zalcman SS. 2004. Permeability of the mouse blood-brain barrier to murine interleukin-2: predominance of a saturable efflux system. Brain Behav Immun 18:434–442
31. Ghersi-Egea JF, Gorevic PD, Ghiso J, Frangione B, Patlak CS, Fenstermacher JD. 1996. Fate of cerebrospinal fluid-borne amyloid β-peptide: rapid clearance into blood and appreciable accumulation by cerebral arteries. J Neurochem 67:880–883
32. Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, Holtzman DM, Miller CA, Strickland DK, Ghiso J, Zlokovic BV. 2000. Clearance of Alzheimer's amyloid-β 1–40 peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest 106:1489–1499
33. Kastin AJ, Fasold MB, Zadina JE. 2002. Endomorphins, Met-enkephalin, Tyr-MIF-1 and the P-glycoprotein efflux system. Drug Metab Dispos 30:231–234
34. Broadwell RD, Sofroniew MV. 1993. Serum proteins bypass the blood-brain barrier for extracellular entry to the central nervous system. Exp Neurol 120:245–263
35. Banks WA. 2004. Are the extracellular pathways a conduit for the delivery of therapeutics to the brain? Curr Pharm Design 10:1365–1370
36. Pardridge WM, Mietus LJ. 1979. Transport of steroid hormones through the rat blood-brain barrier. Primary role of albumin-bound protein. J Clin Invest 64:145–154
37. Pardridge WM, Mietus LJ, Frumar AM, Davidson BJ, Judd HL. 1980. Effects of human serum on transport of testosterone and estradiol into rat brain. Am J Physiol239:E103–E108
38. Banks WA, Kastin AJ, Coy DH. 1982. δ Sleep-inducing peptide crosses the blood-brain-barrier in dogs: some correlations with protein binding. Pharmacol Biochem Behav 17:1009–1014
39. Dickson PW, Aldred AR, Marley PD, Bannister D, Schreiber G. 1986. Rat choroid plexus specializes in the synthesis and the secretion of transthyretin (prealbumin). Regulation of tranthyretin synthesis in choroid plexus is indpendent from that in liver. J Biol Chem 261:3475–3478
40. Hobbs CJ, Jones RE, Plymate SR. 1992. The effects of sex hormone binding globulin (SHBG) on testosterone transport into the cerebrospinal fluid. J Steroid Biochem Mol Biol 42:629–635
41. Havelund S, Plum A, Ribel U, Jonassen I, Volund A, Markussen J, Kurtzhals P. 2004. The mechanism of protraction of insulin detemir, a long-acting, acylated analog of human insulin. Pharm Res 21:1498–1504
42. Davies MJ, Derezinski T, Pedersen CB, Clauson P. 2008. Reduced weight gain with insulin detemir compared to NPH insulin is not explained by a reduction in hypoglycemica. Diabetes Technol Ther 10:273–277
43. Fajardo Montañana C, Hernández Herrero C, Rivas Fernández M. 2008. Less weight gain and hypoglycemia with once-daily insulin detemir than NPH insulin in intensification of insulin therapy in overweight Type 2 diabetes patients: the PREDICTIVE BMI clinical trial. Diabet Med 25:916–923
44. Tschritter O, Schafer SA, Klett J, Pfafflin A, Haring HU, Hennige AM, Fritsche A. 2009. Insulin detemir causes increased symptom awareness during hypoglycemia compared to human insulin. Diabetes Obes Metab 11:1017–1026
45. Banks WA, Morley JE, Lynch JL, Lynch KM, Mooradian AD. 2010. Insulin detemir is not transported across the blood-brain barrier. Peptides 31:2284–2288
46. Banks WA, Kastin AJ. 1998. Differential permeability of the blood-brain barrier to two pancreatic peptides: insulin and amylin. Peptides 19:883–889
47. Banks WA, Moinuddin A, Morley JE. 2001. Regional transport of TNF-‡ across the blood-brain barrier in young ICR and young and aged SAMP8 mice. Neurobiol Aging 22:671–676
48. Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM. 1996. Leptin enters the brain by a saturable system independent of insulin. Peptides 17:305–311
49. Maness LM, Banks WA, Zadina JE, Kastin AJ. 1995. Selective transport of blood-borne interleukin-1 α into the posterior division of the septum of the mouse brain. Brain Res 700:83–88
50. Banks WA, Farr SA, La Scola ME, Morley JE. 2001. Intravenous human interleukin-1 α impairs memory processing in mice: dependence on blood-brain barrier transport into posterior division of the septum. J Pharmacol Exp Ther 299:536–541
51. Caro JF, Kolaczynski JW, Nyce MR, Ohannesian JP, Opentanova I, Goldman WH, Lynn RB, Zhang PL, Sinha MK, Considine RV. 1996. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet 348:159–161
52. Halaas JL, Boozer C, Blair-West J, Fidahusein N, Denton DA, Friedman JM. 1997. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci USA 94:8878–8883
53. Van Heek M, Compton DS, France CF, Tedesco RP, Fawzi AB, Graziano MP, Sybertz EJ, Strader CD, Davis HR., Jr 1997. Diet-induced obese mice develop peripheral, but not central, resistance to leptin. J Clin Invest 99:385–390
54. Banks WA, Coon AB, Robinson SM, Moinuddin A, Shultz JM, Nakaoke R, Morley JE. 2004. Triglycerides induce leptin resistance at the blood-brain barrier. Diabetes 53:1253–1260
55. Kastin AJ, Pan W. 2000. Dynamic regulation of leptin entry into brain by the blood-brain barrier. Regul Pept 92:37–43
56. Kastin AJ, Akerstrom V. 2001. Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice. Neuroendocrinology 73:237–242
57. Kastin AJ, Akerstrom V, Maness LM. 2001. Chronic loss of ovarian function decreases transport of leptin into mouse brain. Neurosci Lett 310:69–71
58. Daubert DL, Chung MY, Brooks VL. 2007. Insulin resistance and impaired baroreflex gain during pregnancy. Am J Physiol Regul Integr Comp Physiol 292:R2188–R2195
59. Azar AS, Brooks VL. 2011. Impaired baroreflex gain during pregnancy in concscious rats: role of brain insulin. Hypertension 57:283–288
60. Daniel PM, Love ER, Moorhouse SR, Pratt OE. 1981. The effect of insulin upon the influx of tryptophan into the brain of the rabbit. J Physiol 312:551–562
61. Cangiano C, Cardelli-Cangiano P, Cascino A, Patrizi MA, Barberini F, Rossi Fanelli F, Capocaccia L, Strom R. 1983. On the stimulation by insulin of tryptophan transport across the blood-brain barrier. Biochem Int 7:617–627
62. Ayre SG, Skaletski B, Mosnaim AD. 1989. Blood-brain barrier passage of azidothymidine in rats: effect of insulin. Res Commun Chem Pathol Pharmacol 63:45–52
63. Catalán RE, Martinez AM, Aragonés MD, Miguel BG, Robles A. 1988. Insulin action on brain microvessels; effect on alkaline phosphatase. Biochem Biophys Res Commun 150:583–590
64. Langston JW, Li W, Harrison L, Aw TY. 2011. Activation of promoter activity of the catalytic subunit of γ-glutamylcysteine ligase (GCL) in brain endothelial cells by insulin requires antioxidant response element 4 and altered glycemic status: implication for GCL expression and GSH synthesis. Free Radic Biol Med 51:1749–1757
65. O'Donnell ME, Lam TI, Tran LQ, Foroutan S, Anderson SE. 2006. Estradiol reduces activity of the blood-brain barrier Na-K-Cl cotransporter and decreases edema formation in permenent middle cerebral artery occlusion. J Cereb Blood Flow Metab 26:1234–1249
66. Fleegal-DeMotta MA, Doghu S, Banks WA. 2009. Angiotensin II modulates BBB permeability via activation of the AT1 receptor in brain endothelial cells. J Cereb Blood Flow Metab 29:640–647
67. Banks WA. 2001. Enhanced leptin transport across the blood-brain barrier by α1-adrenergic agents. Brain Res 899:209–217
68. Urayama A, Grubb JH, Banks WA, Sly WS. 2007. Epinephrine enhances lysosomal enzyme delivery across the blood-brain barrier by up-regulation of the mannose 6-phosphate receptor. Proc Natl Acad Sci USA 104:12873–12878
69. Bauer B, Hartz AM, Miller DS. 2007. Tumor necrosis factor α and endothelin-1 increase P-glycoprotein expression and transport activity at the blood-brain barrier. Mol Pharmacol 71:667–675
70. Hartz AM, Bauer B, Fricker G, Miller DS. 2004. Rapid regulation of P-glycoprotein at the blood-brain barrier by endothelin-1. Mol Pharmacol 66:387–394
71. Reyes TM, Fabry Z, Coe CL. 1999. Brain endothelial cell production of a neuroprotective cytokine, interleukin-6, in response to noxious stimuli. Brain Res 851:215–220
72. Fabry Z, Fitzsimmons KM, Herlein JA, Moninger TO, Dobbs MB, Hart MN. 1993. Production of the cytokines interleukin 1 and 6 by murine brain microvessel endothelium and smooth muscle pericytes. J Neuroimmunol 47:23–34
73. Macvilay S, Fabry Z. 1997. TGF·β cytokine production by murine brain microvessel endothelial cells. Neural Notes 3:21–23
74. Vadeboncoeur N, Segura M, Al-Numani D, Vanier G, Gottschalk M. 2003. Proinflammatory cytokine and chemokine release by human brain microvascular endothelial cells stimulated by Streptococcus suis serotype 2. FEMS Immunol Med Microbiol 35:49–58
75. Pan W, Yu C, Hsuchou H, Khan RS, Kastin AJ. 2009. Cerebral microvascular IL-15 is a novel mediator of TNF action. J Neurochem 111:819–827
76. Faraci FM, Heistad DD. 1998. Regulation of the cerebral circulation: role of endothelium and potassium channels. Physiol Rev 78:53–97
77. Kis B, Kaiya H, Nishi R, Deli MA, Abrahám CS, Yanagita T, Isse T, Gotoh S, Kobayashi H, Wada A, Niwa M, Kangawa K, Greenwood J, Yamashita H, Ueta Y. 2002. Cerebral endothelial cells are a major source of adrenomedullin. J Neuroendocrinol 14:283–293
78. Mándi Y, Ocsovszki I, Szabo D, Nagy Z, Nelson J, Molnar J. 1998. Nitric oxide production and MDR expression by human brain endothelial cells. Anticancer Res18:3049–3052
79. McGuire TR, Trickler WJ, Hock L, Vrana A, Hoie EB, Miller DW. 2003. Release of prostaglandin E-2 in bovine brain endothelial cells after exposure to three unique forms of the antifungal drug amphotericin-B: role of COX-2 in amphotericin-B induced fever. Life Sci 72:2581–2590
80. Didier N, Banks WA, Créminon C, Dereuddre-Bosquet N, Mabondzo A. 2002. HIV-1-induced production of endothelin-1 in an in vitro model of the human blood-brain barrier. Neuroreport 13:1179–1183
81. Verma S, Nakaoke R, Dohgu S, Banks WA. 2006. Release of cytokines by brain endothelial cells: a polarized response to lipopolysaccharide. Brain Behav Immun20:449–455
82. Spranger J, Verma S, Göhring I, Bobbert T, Seifert J, Sindler AL, Pfeiffer A, Hileman SM, Tschöp M, Banks WA. 2006. Adiponectin does not cross the blood-brain barrier, but modifies cytokine expression of brain endothelial cells. Diabetes 55:141–147
83. Mayhan WG, Heistad DD. 1985. Permeability of blood-brain barrier to various sized molecules. Am J Physiol 248:H712–H718
84. Vaughan CJ, Delanty N. 2000. Hypertensive emergencies. Lancet 356:411–417
85. Cao C, Matsumura K, Yamagata K, Watanabe Y. 1997. Involvement of cyclooxygenase-2 in LPS-induced fever and regulation of its mRNA by LPS in the rat brain. Am J Physiol 272:R1712–R1725
86. Inoue W, Matsumura K, Yamagata K, Takemiya T, Shiraki T, Kobayashi S. 2002. Brain-specific endothelial induction of prostaglandin E(2) synthesis enzymes and its temporal relation to fever. Neurosci Res 44:51–61
87. Matsumura K, Cao C, Ozaki M, Morii H, Nakadate K, Watanabe Y. 1998. Brain endothelial cells express cyclooxygenase-2 during lipopolysaccharide-induced fever: light and electron microscopic immunocytochemical studies. J Neurosci 18:6279–6289
88. Kalaria RN, Harik SI. 1989. Increased α 2- and β 2-adrenergic receptors in cerebral microvessels in Alzheimer disease. Neurosci Lett 106:233–238
89. Banks WA, Kastin AJ. 1993. Physiological consequences of the passage of peptides across the blood-brain barrier. Rev Neurosci 4:365–372
90. Ajaya B, Haranath PS. 1982. Effects of insulin administered into cerebrospinal fluid spaces on blood glucose in unanaesthetized and anaesthestized dogs. Indian J Med Res 75:607–615
91. Woods SC, Porte D., Jr 1983. The role of insulin as a satiety factor in the central nervous system. In: Szabo AJ, editor. , ed. Advances in metabolic disorders and CNS regulation of carbohydrate metabolism. Vol 10 New York: Academic Press; 457–482
92. Florant GL, Singer L, Scheurink AJ, Park CR, Richardson RD, Woods SC. 1991. Intraventricular insulin reduces food intake and body weight of marmots during the summer feeding period. Physiol Behav 49:335–338
93. Woods SC, Figlewicz Latemann DP, Schwartz MW, Porte D., Jr 1990. A re-assessment of the regulation of adiposity and appetite by the brain insulin system. Int J Obesity 14(Suppl 3):69–76
94. Van Ree JM, Gaffori O, De Wied D. 1983. In rats, the behavioral profile of CCK-8 related peptides resembles that of antipsychotic agents. Eur J Pharmacol 93:63–78
95. Greeley GH, Jr, Cooper CW, Jeng YJ, Eldridge JC, Thompson JC. 1989. Intracerebroventricular administration of calcitonin enhances glucose-stimulated release of insulin. Regul Pept 24:259–268
96. Drust EG, Crawford IL. 1983. Comparison of the effects of TRH and D-Ala 2 -metenkephalinamide on hippocampal electrical activity and behavior in the unanesthetized rat. Peptides 4:239–243
97. Yehuda S, Youdim MB. 1981. Effects of TRH and PS-24 on colonic temperature and motor activity of rats: possible role of dopamine. Peptides 2:131–135
98. Katsuura G, Yoshikawa K, Itoh S, Hsiao S. 1984. Behavioral effects of thyrotropin releasing hormone in frontal decorticated rats. Peptides 5:899–903
99. Bayliss WM, Starling EH. 1902. The mechanisms of pancreatic secretion. J Physiol 28:325–353
100. Myers K, Goulet M, Rusche J, Boismenu R, Davis M. 2004. Inhibition of fear potentiated startle in rats following peripheral administration of secretin. Psychopharmacol 172:94–99
101. Charlton CG, Miller RL, Crawley JN, Handelmann GE, O'Donohue TL. 1983. Secretin modulation of behavioral and physiological functions in the rat. Peptides4:739–742
102. Mutt V, Carlquist M, Tatemoto K. 1979. Secretin-like bioactivity in extracts of porcine brain. Life Sci 25:1703–1707
103. O'Donohue TL, Charlton CG, Miller RL, Boden G, Jacobowitz DM. 1981. Identification, characterization, and distribution of secretin immunoreactivity in rat and pig brain. Proc Natl Acad Sci USA 78:5221–5224
104. Kopin AS, Wheeler MB, Leiter AB. 1990. Secretin: structure of the precursor and tissue distribution of the mRNA. Proc Natl Acad Sci USA 87:2299–2303
105. Banks WA, Goulet M, Rusche J, Niehoff ML, Boismenu R. 2002. Differential transport of secretin analog across the blood-brain and blood-cerebrospinal fluid barriers of the mouse. J Pharmacol Exp Ther 302:1062–1069
106. Fremeau RT, Jr, Jensen RT, Charlton CG, Miller RL, O'Donohue TL, Moody TW. 1983. Secretin: specific binding to rat brain membranes. J Neurosci 3:1620–1625
107. Ishihara T, Nakamura S, Kaziro Y, Takahashi T, Takahashi K, Nagata S. 1991. Molecular cloning and expression of a cDNA encoding the secretin receptor. EMBO J10:1635–1641
108. Karelson E, Laasik J, Sillard R. 1995. Regulation of adenylate cyclase by galanin, neuropeptide Y, secretin and vasoactive intestinal polypeptide in rat frontal cortex, hippocampus and hypothalamuc. Neuropeptides 28:21–28
109. Yung WH, Leung PS, Ng SS, Zhang J, Chan SC, Chow BK. 2001. Secretin facilitates GABA transmission in the cerebellum. J Neurosci 21:7063–7068
110. Conter RL, Hughes MT, Kauffman GL., Jr 1996. Intracerebroventricular secretin enhances pancreatic volume and bicarbonate response in rats. Surgery 119:208–213
111. Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, Aguilar-Bryan L, Rossetti L. 2005. Hypothalmic K(ATP) channels control hepatic glucose production. Nature 434:1026–1031
112. Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, Knapp DJ, Crews FT. 2007. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. GLIA 55:453–462
113. Bishop PM, De Mowbray RR. 1951. Pseudohypoparathyroidism Proc R Soc Med 44:952–954
114. Verhoeven GF, Wilson JD. 1979. The syndromes of primary hormone resistance. Metabolism 28:253–289
115. Refetoff S. 1982. Syndromes of thyroid hormone resistance. Am J Physiol 243:E88–E98
116. Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O'Kirwan F, Whitby R, Liang L, Cohen P, Bhasin S, Krauss RM, Veldhuis JD, Wagner AJ, DePaoli AM, McCann SM, Wong ML. 2004. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci USA 101:4531–4536
117. Pelleymounter MA, Cullen MJ, Baker MB, Hecht R, Winters D, Boone T, Collins F. 1995. Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269:540–543
118. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, Lubina JA, Patane J, Self B, Hunt P, McCamish M. 1999. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA 282:1568–1575
119. El-Haschimi K, Pierroz DD, Hileman SM, Bjørbaek C, Flier JS. 2000. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obestiy. J Clin Invest 105:1827–1832
120. Banks WA, LeBel C. 2002. Strategies for the delivery of leptin to the CNS. J Drug Target 10:297–308
121. Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M, Porte D., Jr 1998. Cerebrosinal fluid and plasma insulin levels in Alzheimer's disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology 50:164–168
122. Frolich L, Blum-Degen D, Bernstein HG, Engelsberger S, Humrich J, Laufer S, Muschner D, Thalheimer A, Turk A, Hoyer S, Zochling R, Boissl KW, Jellinger K, Riederer P. 1998. Brain insulin and insulin receptors in aging and sporadic Alzheimer's disease. J Neural Transm 105:423–428
123. Hoyer S. 1998. Is sporadic Alzheimer's disease the brain type of non-insulin dependent diabetes mellitus? A challenging hypothesis. J Neural Transm 105:415–422
124. Craft S, Asthana S, Schellenberg G, Cherrier M, Baker LD, Newcomer J, Plymate S, Latendresse S, Petrova A, Raskind M, Peskind E, Lofgreen C, Grimwood K. 1999. Insulin metabolism in Alzheimer's disease differs according to apolipoprotein E genotype and gender. Neuroendocrinology 70:146–152
125. Watson GS, Craft S. 2003. The role of insulin resistance in the pathogenesis of Alzheimer's disease: implications for treatment. CNS Drugs 17:27–45
126. Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, Kern W. 2004. Intranasal insulin improves memory in humans. Psychoneuroendocrinology29:1326–1334
127. Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, Plymate SR, Cherrier MM, Schellenberg GD, Frey WH, II, Craft S. 2008. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-β in memory-impaired adults. J Alzheimers Dis 13:323–331
128. Craft S, Newcomer J, Kanne S, Dagogo-Jack S, Cryer P, Sheline Y, Luby J, Dagogo-Jack A, Alderson A. 1996. Memory improvement following induced hyperinsulinemia in Alzheimer's disease. Neurobiol Aging 17:123–130
129. Craft S, Asthana S, Newcomer JW, Wilkinson CW, Matos IT, Baker LD, Cherrier M, Lofgreen C, Latendresse S, Petrova A, Plymate S, Raskind M, Grimwood K, Veith RC. 1999. Enhancement of memory in Alzheimer disease with insulin and somatostatin, but not glucose. Arch Gen Psychiatry 56:1135–1140
130. Hoyer S. 2004. Modulation of memory by insulin and glucose: neuropsychological observations in Alzheimer's disease. Eur J Pharmacol 490:115–125
131. Banks WA, DiPalma CR, Farrell CL. 1999. Impaired transport of leptin across the blood-brain barrier in obesity. Peptides 20:1341–1345
132. Huber JD, VanGilder RL, Houser KA. 2006. Streptozotocin-induced diabetes progressively increases blood-brain barrier permeability in specific brain regions in rats. Am J Physiol Heart Circ Physiol 291:H2660–H2668
133. Opp MR, Krueger JM. 1991. Interleukin 1-receptor antagonist blocks interleukin 1-induced sleep and fever. Am J Physiol 260:R453–R457


Figures and Tables



Fig. 1.

An external file that holds a picture, illustration, etc.Object name is zee0091266430001.jpg


Mechanisms for translocation across the BBB of hormones and their messages between the brain and blood. The role of barrier prevents substances from crossing the BBB as exemplified by steroids that are protein bound. Permeability of the BBB is often by way of transmembrane diffusion as exemplified by unbound steroid hormone or by way of transporters as exemplified by leptin. The cells forming the BBB are themselves targets of circulating substances as exemplified by the many effects that insulin has on brain endotheial cell function. The cells that form the BBB are themselves endocrine-like in that they can secret substances into the blood or into the brain as exemplified by IL-6 secretion by brain endothelial cells. Relay is a special feature arising from the unique separation of brain endothelial cells into luminal and abluminal membranes and allows the BBB cell to receive an input on one membrane and to secrete from the other as exemplified here by adiponectin acting at the luminal membrane to alter secretion of IL-6 from the abluminal membrane. Extracellular pathways (data not shown) can be used by albumin and by very large and stable proteins (antibodies, erythropoietin) to gain access to the CNS.






Table 1.


Endocrinology and the BBB: examples of disease associations



Disease/condition Contribution References
Obesity Defective blood-to-brain transport of leptin (131)
Diabetes mellitus Disruption of the BBB (132)
Hypertensive urgency/emergency Angiotensin II induction of macropinocytosis at BEC (66)
Baroreceptor deficiency in pregnancy Decreased insulin in CNS (59)
Alzheimer's disease Insulin resistance in brain (125)
Fever (sickness behavior) Prostaglandin secretion in response to IL-1 (133)






Fig. 2.

An external file that holds a picture, illustration, etc.Object name is zee0091266430002.jpg


Hormones crossing the BBB can modulate their peripheral affects. A hormone crossing the BBB can reinforce its peripheral actions by inducing its release from central stores (illustrated in the left panel by TNF) or by other mechanisms such as vagal relay (illustrated in the middle panel by insulin action on gluconeogenesis). A hormone crossing the BBB can attenuate its peripheral actions as exemplified in the right panel by insulin's effects on feeding.