Vous êtes sur la page 1sur 11

Section I

Hormones and
Hormone Action

1
CHAPTER 1 
Principles of Endocrinology
HENRY M. KRONENBERG • SHLOMO MELMED • P. REED LARSEN • KENNETH S. POLONSKY

The Evolutionary Perspective, 2 the endocrine and nervous systems as two distinct mecha-
Endocrine Glands, 4 nisms for coordination and control of organ function.
Thus, endocrinology found its first home in the discipline
Transport of Hormones in Blood, 5 of mammalian physiology.
Target Cells as Active Participants, 6 Work over the next several decades by biochemists,
Control of Hormone Secretion, 7 physiologists, and clinical investigators led to the charac-
terization of many hormones secreted into the bloodstream
Hormone Measurement, 9
from discrete glands or other organs. These investigators
Endocrine Diseases, 9 showed that diseases such as hypothyroidism and diabetes
Diagnostic and Therapeutic Uses of Hormones, 10 could be treated successfully for the first time by replacing
What We Don’t Know (Yet), 11 specific hormones. These initial triumphs formed the foun-
dation of the clinical specialty of endocrinology.
Advances in cell biology, molecular biology, and genet-
ics over the ensuing years began to explain the mecha-
KEY  POINTS
nisms of endocrine diseases and of hormone secretion and
action. Even though these advances have embedded endo-
• Endocrinology is a scientific and medical discipline
crinology in the framework of molecular cell biology, they
with a unique focus on hormones and features a
have not changed the essential subject of endocrinology—
multidisciplinary approach to understanding hormones
the signaling that coordinates and controls the functions
and their diseases.
of multiple organs and processes. Herein we survey the
• Endocrine and paracrine systems differ in important
general themes and principles that underpin the diverse
respects that illustrate the evolutionary pressures on
approaches used by clinicians, physiologists, biochemists,
these distinct cell signaling strategies.
cell biologists, and geneticists to understand the endo-
• Hormone-secreting cells are designed to efficiently
crine system.
synthesize hormones and secrete them in a regulated
way.
• Hormones in the bloodstream often are associated with
binding proteins to allow their solubility, keep them from THE EVOLUTIONARY PERSPECTIVE
degradation and renal excretion, and regulate their
stability in the extracellular space. Hormones can be defined as chemical signals secreted into
the bloodstream that act on distant tissues, usually in a
• Hormones either act on receptors on the plasma
regulatory fashion. Hormonal signaling represents a special
membranes of target cells or move into cells to bind to
case of the more general process of signaling between cells.
intracellullar receptors; in either case, the target cell is
Even unicellular organisms, such as baker’s yeast, Saccharo-
not a passive recipient of signals but rather has key roles
myces cerevisiae, secrete short peptide mating factors that
in regulating the responses to hormones.
act on receptors of other yeast cells to trigger mating
• Control of hormone secretion involves multiple inputs
between the two cells. These receptors resemble the ubiq-
from distant targets, nervous system inputs, and local
uitous family of seven membrane-spanning mammalian
paracrine and autocrine factors, all leading to complex
receptors that respond to ligands as diverse as photons
patterns of circadian secretion, pulsatile secretion,
and glycoprotein hormones. Because these yeast receptors
secretion driven by homeostatic stimuli, or stimuli that
trigger activation of heterotrimeric G proteins just as mam-
lead to secular changes over the lifespan.
malian receptors do, this conserved signaling pathway
• Endocrine diseases fall into broad categories of hormone
must have been present in the common ancestor of yeast
over- or underproduction, altered tissue response to
and humans.
hormones, or tumors arising from endocrine tissue.
Signals from one cell to adjacent cells, so-called para-
• Hormones and synthetic molecules designed to interact
crine signals, often use the same molecular pathways used
with hormone receptors are administered to diagnose
by hormonal signals. For example, the sevenless receptor
and treat diseases.
controls the differentiation of retinal cells in the Drosophila
eye by responding to a membrane-anchored signal from
an adjacent cell. Sevenless is a membrane-spanning recep-
About a hundred years ago, Starling coined the term tor with an intracellular tyrosine kinase domain that
hormone to describe secretin, a substance secreted by the signals in a way that closely resembles the signaling by
small intestine into the bloodstream to stimulate pancre- hormone receptors such as the insulin receptor tyrosine
atic secretion. In his Croonian Lectures, Starling considered kinase. Because paracrine factors and hormones can share

2
CHAPTER 1  Principles of Endocrinology 3

Regulation of signaling: endocrine


Source: gland Distribution: bloodstream Non-target organ
• No contribution to • Universal — almost • Metabolism
specificity of target • Importance of dilution
• Synthesis/secretion

Target cell
• Receptor: source of specificity
• Responsiveness:
Number of receptors
Downstream pathways
Other ligands
Metabolism of ligand/receptor
All often regulated by ligand

Regulation of signaling: paracrine


Source: adjacent cell Target cell
• Major determinant of target • Receptor:
• Synthesis/secretion Specificity and sensitivity
Diffusion barrier
Determinant of gradient
• Induced inhibitory pathways,
ligands, and binding proteins

Distribution: matrix
• Diffusion distance
• Binding proteins: BMP, IGF Figure 1-1 Comparison of determinants of endocrine and paracrine
• Proteases signaling. BMP, bone morphogenetic protein; IGF, insulin-like growth
• Matrix components factor.

signaling machinery, it is not surprising that hormones systems (Fig. 1-1). Paracrine signals do not travel very far;
can, in some settings, act as paracrine factors. Testosterone, consequently, the specific site of origin of a paracrine factor
for example, is secreted into the bloodstream but also acts determines where it will act and provides specificity to that
locally in the testes to control spermatogenesis. Insulin-like action. When the paracrine factor bone morphogenetic
growth factor 1 (IGF-1) is a polypeptide hormone secreted protein 4 (BMP4) is secreted by cells in the developing
into the bloodstream from the liver and other tissues but kidney, BMP4 regulates the differentiation of renal cells;
is also a paracrine factor made locally in most tissues when the same factor is secreted by cells in bone, it regu-
to control cell proliferation. Furthermore, one receptor lates bone formation. Thus, the site of origin of BMP4
can mediate actions of a hormone, such as parathyroid determines its physiologic role. In contrast, because hor-
hormone (PTH), and of a paracrine factor, such as parathy- mones are secreted into the bloodstream, their sites of
roid hormone–related protein. In some cases, the paracrine origin are often divorced from their functions. Like BMP4,
actions of “hormones” have functions quite unrelated to thyroid hormone, for example, acts in many tissues, but
the hormonal functions. For example, macrophages syn- the site of origin of thyroid hormone in a gland in the neck
thesize the active form of vitamin D, 1,25-dihydroxyvitamin has nothing to do with the sites of action of the hormone.
D3 (1,25[OH]2D3), which can then bind to vitamin D recep- Because the specificity of paracrine factor action is so
tors in the same cells and stimulate production of antimi- dependent on its precise site of origin, elaborate mecha-
crobial peptides.1 The vitamin D 1α-hydroxylase responsible nisms have evolved to regulate and constrain the diffusion
for activating 25-hydroxyvitamin D is synthesized in mul- of paracrine factors. Paracrine factors of the hedgehog
tiple tissues in which it has functions unrelated to the family, for example, are covalently bound to cholesterol to
calcium homeostatic actions of the 1,25(OH)2D3 hormone. constrain the diffusion of these molecules in the extracel-
One can speculate that the hormonal actions of vitamin D lular milieu. Most paracrine factors interact with binding
might have evolved well after the paracrine vitamin D proteins that block their action and control their diffusion.
system provided the raw materials for the hormonal system. Chordin, noggin, and many other distinct proteins all bind
Target cells respond similarly to signals that reach them to various members of the BMP family to regulate their
from the bloodstream (hormones) or from the cell next action, for example. Proteases such as tolloid then destroy
door (paracrine factors); the cellular response machinery the binding proteins at specific sites to liberate BMPs so
does not distinguish the sites of origin of signals. The that they can act on appropriate target cells.
shared final common pathways used by hormonal and Hormones have rather different constraints. Because
paracrine signals should not, however, obscure important they diffuse throughout the body, they must be synthe-
differences between hormonal and paracrine signaling sized in enormous amounts relative to the amounts
4 SECTION I  Hormones and Hormone Action

of paracrine factors needed at specific locations. This syn- hormones is that they are synthesized in discrete glands
thesis usually occurs in specialized cells designed for that and have no function other than activating receptors on
specific purpose. Hormones must then be able to travel in cell membranes or in the nucleus. Cholesterol, which is
the bloodstream and diffuse in effective concentrations converted in cells to oxygenated derivatives that activate
into tissues. Therefore, for example, lipophilic hormones the LXR receptor, in contrast, uses a hormonal strategy to
bind to soluble proteins that allow them to travel in the regulate its own metabolism. Other orphan nuclear recep-
aqueous media of blood at relatively high concentrations. tors similarly respond to ligands such as bile acids and fatty
The ability of hormones to diffuse through the extracel- acids. These “hormones” have important metabolic roles
lular space means that the local concentration of hormone quite separate from their signaling properties, although the
at target sites will rapidly decrease when glandular secre- hormone-like signaling serves to allow regulation of the
tion of the hormone stops. Because hormones diffuse metabolic function. The calcium-sensing receptor is an
throughout extracellular fluid quickly, hormonal metabo- example from the G protein–linked receptor family that
lism can occur in specialized organs such as the liver and responds to a nonclassic ligand, ionic calcium. Calcium is
kidney in a manner that determines the effective hormone released into the bloodstream from bone, kidney, and
concentration in other tissues. intestine and acts on the calcium-sensing receptor on para-
Thus, paracrine factors and hormones use several dis- thyroid cells, renal tubular cells, and other cells to coordi-
tinct strategies to control their biosynthesis, sites of nate cellular responses to calcium. Thus, many important
action, transport, and metabolism. These differing strate- metabolic factors have taken on hormonal properties as
gies probably explain partly why a hormone such as IGF-1, part of a regulatory strategy.
unlike its close relative, insulin, has multiple binding pro-
teins that control its action in tissues. IGF-1 exhibits a
double life as both a hormone and a paracrine factor. Pre- ENDOCRINE GLANDS
sumably, the local actions of IGF-1 mandate an elaborate
binding protein apparatus to enable appropriate hormone Hormone formation may occur either in localized collec-
signaling. tions of specific cells, the endocrine glands, or in cells that
All the major hormonal signaling programs—G protein– have additional roles. Many protein hormones, such as
coupled receptors, tyrosine kinase receptors, serine/ growth hormone (GH), PTH, prolactin (PRL), insulin, and
threonine kinase receptors, ion channels, cytokine recep- glucagon, are produced in dedicated cells by standard
tors, nuclear receptors—are also used by paracrine factors. protein synthetic mechanisms common to all cells. These
In contrast, several paracrine signaling programs are used secretory cells usually contain specialized secretory gran-
only by paracrine factors and probably not by hormones. ules designed to store large amounts of hormone and to
For example, Notch receptors respond to membrane-based release the hormones in response to specific signals. Forma-
ligands to control cell fate, but no blood-borne ligands use tion of small hormone molecules initiates with commonly
Notch-type signaling (at least, none is currently known). found precursors, usually in specific glands such as the
Perhaps the intracellular strategy used by Notch, which adrenals, gonads, or thyroid. In the case of the steroid
involves cleavage of the receptor and subsequent nuclear hormones, the precursor is cholesterol, which is modified
actions of the receptor’s cytoplasmic portion, is too inflex- by various hydroxylations, methylations, and demethyl-
ible to serve the purposes of hormones. ations, all using cytochrome P450-based reactions to form
The analyses of the complete genomes of multiple bac- the glucocorticoids, androgens, estrogens, and their bio-
terial species, the yeast S. cerevisiae, the fruit fly Drosophila logically active derivatives.
melanogaster, the worm Caenorhabditis elegans, the plant However, not all hormones are formed in dedicated and
Arabidopsis thaliana, humans, and many other species have specialized endocrine glands. For example, the protein
allowed a comprehensive view of the signaling machinery hormone leptin, which regulates appetite and energy
used by various forms of life. As noted already, S. cerevisiae expenditure, is formed in adipocytes, thus providing a spe-
uses G protein–linked receptors; this organism, however, cific signal reflecting the nutritional state of the organism
lacks tyrosine kinase receptors and nuclear receptors that to the central nervous system. The cholesterol derivative,
resemble the estrogen/thyroid receptor family. In contrast, 7-dehydrocholesterol, the precursor of vitamin D, is pro-
the worm and fly share with humans the use of each of duced in skin keratinocytes by a photochemical reaction.
these signaling pathways, although with substantial varia- The enteroendocrine system comprises a unique hormonal
tion in numbers of genes committed to each pathway. For system in which peptide hormones that regulate metabolic
example, the Drosophila genome encodes 20 nuclear recep- and other responses to oral nutrients are produced and
tors, the C. elegans genome encodes 270, and the human secreted by specialized endocrine cells scattered through-
genome encodes 48. These patterns suggest that ancient out the intestinal epithelium.
multicellular animals must have already established the Thyroid hormone synthesis occurs via a unique pathway.
signaling systems that are the foundation of the endocrine The thyroid cell synthesizes a 660,000-kDa homodimer,
system as we know it in mammals. thyroglobulin, which is then iodinated at specific iodoty-
Even before the sequencing of the human genome, rosines. Certain of these “couple” to form the iodothyro-
sequence analyses had made clear that many receptor nine molecule within thyroglobulin, which is then stored
genes are found in mammalian genomes for which no clear in the lumen of the thyroid follicle. In order for this to
ligand or function was known. The analyses of these occur, the thyroid cell must concentrate the trace quanti-
“orphan” receptors have succeeded in broadening the ties of iodide from the blood and oxidize it via a specific
current understanding of hormonal signaling. For example, peroxidase. Release of thyroxine (T4) from the thyroglobu-
the liver X receptor (LXR) was one such orphan receptor lin requires its phagocytosis and cathepsin-catalyzed diges-
found when searching for unknown nuclear receptors. Sub- tion by the same cells.
sequent experiments showed that oxygenated derivatives Hormones are synthesized in response to biochemical
of cholesterol are the ligands for LXR, which regulates signals generated by various modulating systems. Many of
genes involved in cholesterol and fatty acid metabolism.2 these systems are specific to the effects of the hormone
The examples of LXR and many others raise the question product; for example, PTH synthesis is regulated by the
of what constitutes a hormone. The classic view of concentration of ionized calcium. Insulin synthesis is
CHAPTER 1  Principles of Endocrinology 5

regulated by the concentration of glucose. For others, such clinical and laboratory consequences. Recognition and cor-
as gonadal, adrenal, and thyroid hormones, control of rection of these are the essence of clinical endocrinology.
hormone synthesis is achieved by the hormonostatic func-
tion of the hypothalamic-pituitary axis. Cells in the hypo-
thalamus and pituitary monitor the circulating hormone TRANSPORT OF HORMONES IN BLOOD
concentration and secrete trophic hormones, which acti-
vate specific pathways for hormone synthesis and release. Protein hormones and some small molecules, such as the
Typical examples are luteinizing hormone, follicle- catecholamines, are water-soluble and are readily trans-
stimulating hormone, thyroid-stimulating hormone, and ported via the circulatory system. Others are nearly insol-
adrenocorticotropic hormone (LH, FSH, TSH, and ACTH, uble in water (e.g., the steroid and thyroid hormones) and
respectively). their distribution presents special problems. Such mole-
These trophic hormones increase rates of hormone syn- cules are bound to 50- to 60-kDa carrier plasma glycopro-
thesis and secretion, and they may induce target cell divi- teins such as thyroxine-binding globulin (TBG), sex
sion, thus causing enlargement of the various target glands. hormone–binding globulin (SHBG), and corticosteroid-
For example, in hypothyroid individuals living in iodine- binding globulin (CBG) as well as to albumin. These ligand-
deficient areas of the world, TSH secretion causes a marked protein complexes serve as reservoirs of these hormones,
hyperplasia of thyroid cells. In such regions, the thyroid ensure ubiquitous distribution of their water-insoluble
gland may be 20 to 50 times its normal size. Adrenal hyper- ligands, and protect the small molecules from rapid inac-
plasia occurs in patients with genetic deficiencies in corti- tivation or excretion in the urine or bile. The protein-
sol formation. Hypertrophy and hyperplasia of parathyroid bound hormones exist in rapid equilibrium with the
cells, in this case initiated by an intrinsic response to the often-minute quantities of hormone in the aqueous plasma.
stress of hypocalcemia, occur in patients with renal insuf- It is this “free” fraction of the circulating hormone that is
ficiency or calcium malabsorption. taken up by the target cell. It has been shown, for example,
Hormones may be fully active when released into the that if tracer thyroid hormone is injected into the portal
bloodstream (e.g., GH or insulin), or they may require vein in a protein-free solution, it is bound to hepatocytes
activation in specific cells to produce their biologic effects. at the periphery of the hepatic sinusoid. When the same
These activation steps are often highly regulated. For experiment is repeated with a protein-containing solution,
example, the T4 released from the thyroid cell is a prohor- there is a uniform distribution of the tracer hormone
mone that must undergo a specific deiodination to form throughout the hepatic lobule.3 Despite the very high
the active 3,5,3′-triiodothyronine (T3). This deiodination affinity of some of the binding proteins for their ligands,
reaction can occur in target tissues, such as in the central one specific protein may not be essential for hormone
nervous system; in the thyrotrophs, where T3 provides distribution. For example, in humans with a congenital
feedback regulation of TSH production; or in hepatic and deficiency of TBG, other proteins, transthyretin (TTR) and
renal cells, from which it is released into the circulation for albumin, subsume its role. Because the affinity of these
uptake by all tissues. A similar postsecretory activation step secondary thyroid hormone transport proteins is several
catalyzed by a 5α-reductase causes tissue-specific activation orders of magnitude lower than that of TBG, it is possible
of testosterone to dihydrotestosterone in target tissues, for the hypothalamic-pituitary feedback system to main-
including the male urogenital tract and genital skin as well tain free thyroid hormone in the normal range at a much
as in liver. Vitamin D undergoes hydroxylation at the 25 lower total hormone concentration. The fact that the free
position in the liver and in the 1 position in the kidney. hormone concentration is normal in subjects with TBG
Both hydroxylations must occur to produce the active deficiency indicates that it is this free moiety that is
hormone, 1,25-hydroxyvitamin D. The activity of the defended by the hypothalamic-pituitary axis and is the
1α-hydroxylase, but not the 25-hydroxylase, is stimulated active hormone.4
by PTH and reduced plasma phosphate but is inhibited by The availability of gene targeting techniques has allowed
calcium, 1,25-hydroxyvitamin D, and fibroblast growth specific tests of the physiologic role of several hormone-
factor 23 (FGF23). binding proteins. For example, mice with targeted inactiva-
Hormones are synthesized as required on a daily, hourly, tion of the vitamin D–binding protein (DBP) have been
or minute-to-minute basis with minimal storage, but there generated.5 Although the absence of DBP markedly reduces
are significant exceptions. One is the thyroid gland, which the circulating concentration of vitamin D, the mice are
contains enough stored hormone to last for about 2 otherwise normal. However, they do show enhanced sus-
months. This storage permits a constant supply of this ceptibility to a vitamin D–deficient diet because of the
hormone despite significant variations in the availability reduced reservoir of this sterol. In addition, the absence of
of iodine. However, if iodine deficiency is prolonged, the DBP markedly reduces the half-life of 25-hydroxyvitamin
normal reservoirs of T4 can be depleted. D by accelerating its hepatic uptake, making the mice less
The various feedback signaling systems exemplified susceptible to vitamin D intoxication.
earlier enable the hormonal homeostasis characteristic of In rodents, TTR carries retinol-binding protein and is
virtually all endocrine systems. Regulation may include the also the principal thyroid hormone–binding protein. This
central nervous system or local signal recognition mecha- protein is synthesized in the liver and in the choroid
nisms in the glandular cells, such as the calcium-sensing plexus. It is the major thyroid hormone–binding protein
receptor of the parathyroid cell. Superimposed, centrally in the cerebrospinal fluid of both rodents and humans and
programmed increases and decreases in hormone secretion was thought to perhaps serve an important role in thyroid
or activation through neuroendocrine pathways also occur. hormone transport into the central nervous system. This
Examples include the circadian variation in the secretion hypothesis has been disproved by the fact that mice
of ACTH directing the synthesis and release of cortisol. The without TTR have normal concentrations of T4 in the brain
monthly menstrual cycle exemplifies a system with much as well as free T4 in the plasma.6,7 To be sure, the serum
longer periodicity that requires a complex synergism concentrations of vitamin A and total T4 are decreased, but
between central and peripheral axes of the endocrine the knockout mice have no signs of vitamin A deficiency
glands. Disruption of hormonal homeostasis due to glan- or hypothyroidism. Such studies suggest that these pro-
dular or central regulatory system dysfunction has both teins primarily serve distributive and reservoir functions.
6 SECTION I  Hormones and Hormone Action

Progesterone

O R

O RR O
XTyr

TF P TFTyr P
R ss
PKA s
Figure 1-2 Hormonal signaling by cell-surface and intracellular Target gene
receptors. The receptors for the water-soluble polypeptide hor- IGF-1
XTyr P
mones, luteinizing hormone (LH), and insulin-like growth factor 1 cAMP
(IGF-1) are integral membrane proteins located at the cell surface.
They bind the hormone-utilizing extracellular sequences and trans-
duce a signal by the generation of second messengers: cyclic AC
adenosine monophosphate (cAMP) for the LH receptor and tyrosine- ATP AAAAA
phosphorylated substrates for the IGF-1 receptor. Although effects G mRNAs
on gene expression are indicated, direct effects on cellular proteins R
s ss

(e.g., ion channels) are also observed. In contrast, the receptor for
the lipophilic steroid hormone progesterone resides in the cell LH
nucleus. It binds the hormone and becomes activated and capable
of directly modulating target gene transcription. AC, adenylate Proteins
cyclase; ATP, adenosine triphosphate; G, heterotrimeric G protein;
mRNAs, messenger RNAs; PKA, protein kinase A; R, receptor mol-
ecule; TF, transcription factor; Tyr, tyrosine found in protein X;
X, unknown protein substrate. (From Mayo K. Receptors: molecular
mediators of hormone action. In: Conn PM, Melmed S, eds. Endo-
crinology: Basic and Clinical Principles. Totowa, NJ: Humana Press;
1997:11.) Biological responses

Protein hormones and some small ligands (e.g., cate- selective hormone receptors. Receptor expression thus
cholamines) produce their effects by interacting with cell- determines which cells will respond, as well as the nature
surface receptors. Others, such as the steroid and thyroid of the intracellular effector pathways activated by the
hormones, must enter the cell to bind to cytosolic or hormone signal. Receptor proteins may be localized to the
nuclear receptors. In the past, it has been thought that cell membrane, cytoplasm, or nucleus. Broadly, polypep-
much of the transmembrane transport of hormones was tide hormone receptors are cell membrane–associated, but
passive. Evidence now shows that there are specific trans- steroid hormones selectively bind soluble intracellular pro-
porters involved in cellular uptake of thyroid hormone.7 teins (Fig. 1-2).
This activity may be found to be the case for other small Membrane-associated receptor proteins usually consist
ligands as well, revealing yet another mechanism for of extracellular sequences that recognize and bind ligand,
ensuring the distribution of a hormone to its site of transmembrane-anchoring hydrophobic sequences, and
action. Studies in mice missing megalin, a large, cell- intracellular sequences, which initiate intracellular sig­
surface protein in the low-density lipoprotein (LDL) naling. Intracellular signaling is mediated by covalent
receptor family, suggest that estrogen and testosterone, modification and activation of intracellular signaling mol-
bound to SHBG, use megalin to enter certain tissues ecules (e.g., signal transducers and activators of transcrip-
while still bound to SHBG.8 In this case, therefore, the tion [STAT] proteins) or by generation of small molecule
hormone bound to SHBG, rather than “free” hormone, is second messengers (e.g., cyclic adenosine monophosphate)
the active moiety that enters cells. It is unclear how gen- through activation of heterotrimeric G proteins. Subunits
erally this apparent exception to the “free hormone” of these G proteins (α-, β-, and γ-subunits) activate or sup-
hypothesis occurs. press effector enzymes and ion channels that generate the
second messengers. Some of these receptors may in fact
exhibit constitutive activity and have been shown to signal
in the absence of added ligand.
TARGET CELLS AS ACTIVE Several growth factors and hormone receptors (e.g., for
PARTICIPANTS insulin) behave as intrinsic tyrosine kinases or activate
intracellular protein tyrosine kinases. Ligand activation
Hormones determine cellular target actions by binding may cause receptor dimerization (e.g., GH) or heterodimer-
with high specificity to receptor proteins. Whether or not ization (e.g., interleukin 6), followed by activation of intra-
a peripheral cell is hormonally responsive depends to a cellular phosphorylation cascades. These activated proteins
large extent on the presence and function of specific and ultimately determine specific nuclear gene expression.
CHAPTER 1  Principles of Endocrinology 7

Both the number of receptors expressed per cell, as well TABLE 1-1 
as their responses, are also regulated, thus providing a Diseases Caused by Mutations in G
further level of control for hormone action. Several mecha- Protein–Coupled Receptors
nisms account for altered receptor function. Receptor
endocytosis causes internalization of cell-surface receptors; Condition* Receptor Inheritance Δ Function†
the hormone-receptor complex is subsequently dissoci- Retinitis pigmentosa Rhodopsin AD/AR Loss
ated, resulting in abrogation of the hormone signal. Recep- Nephrogenic diabetes Vasopressin X-linked Loss
tor trafficking may then result in recycling back to the cell insipidus V2
surface (e.g., as for insulin), or the internalized receptor Isolated glucocorticoid ACTH AR Loss
may undergo lysosomal degradation. Both these mecha- deficiency
nisms triggered by activation of receptors effectively lead Color blindness Red/green X-linked Loss
opsins
to impaired hormone signaling by downregulation of these Familial precocious LH AD (male) Gain
receptors. The hormone signaling pathway may also be puberty
downregulated by receptor desensitization (e.g., as for Familial hypercalcemia Ca2+ sensing AD Loss
epinephrine); ligand-mediated receptor phosphorylation Neonatal severe Ca2+ sensing AR Loss
leads to a reversible deactivation of the receptor. Desensi- parathyroidism
tization mechanisms can be activated by a receptor’s ligand Dominant form Ca2+ sensing AD Gain
(homologous desensitization) or by another signal (heter- hypocalcemia
ologous desensitization), thereby attenuating receptor Congenital TSH AD Gain
hyperthyroidism
signaling in the continued presence of ligand. Receptor
Resistance to thyroid TSH AR (comp Loss
function may also be limited by action of specific phospha- hormone het)
tases (e.g., Src homology phosphatase [SHP]) or by intracel- Hyperfunctioning TSH Somatic Gain
lular negative regulation of the signaling cascade (e.g., thyroid adenoma
suppressor of cytokine signaling [SOCS] proteins inhibiting Metaphyseal PTH-PTHrP Somatic Gain
Janus kinase/signal transducers and activators of transcrip- chondrodysplasia
tion [JAK-STAT] signaling). Certain ligand-receptor com- Hirschsprung disease Endothelin-B Multigenic Loss
plexes may also translocate to the nucleus.
Mutational changes in receptor structure can also deter- Coat color alteration MSH AD/AR Loss and
(E locus, mice) gain
mine hormone action. Constitutive receptor activation
Dwarfism (little locus, GHRH AR Loss
may be induced by activating mutations (e.g., TSH recep- mice)
tor) leading to endocrine organ hyperfunction, even in the
absence of hormone. Conversely, inactivating receptor *All are human conditions with the exception of the final two entries, which
mutations may lead to endocrine hypofunction (e.g., tes- refer to the mouse.

Loss of function refers to inactivating mutations of the receptor, and gain of
tosterone or vasopressin receptors). These syndromes are function to activating mutations.
well characterized and are well described in this volume ACTH, adrenocorticotropic hormone; AD, autosomal dominant inheritance;
(Table 1-1). AR, autosomal recessive inheritance; FSH, follicle-stimulating hormone;
The functional diversity of receptor signaling also results GHRH, growth hormone–releasing hormone; LH, luteinizing hormone;
MSH, melanocyte-stimulating hormone; PTH-PTHrP, parathyroid hormone
in overlapping or redundant intracellular pathways. For and parathyroid hormone–related peptide; TSH, thyroid-stimulating
example, both GH and cytokines activate JAK-STAT signal- hormone.
ing, whereas the distal effects of these stimuli clearly differ. From Mayo K. Receptors: molecular mediators of hormone action. In: Conn
Thus, despite common signaling pathways, hormones PM, Melmed S, eds. Endocrinology: Basic and Clinical Principles. Totowa,
NJ: Humana Press; 1997:27.
elicit highly specific cellular effects. Tissue- or cell-type
genetic programs or receptor-receptor interactions at the
cell surface (e.g., dopamine D2 with somatotropin release–
inhibiting factor [SRIF] receptor hetero-oligomerization) Hormone secretion also adheres to rhythmic patterns.
may also confer specific cellular response to a hormone and Circadian rhythms serve as adaptive responses to environ-
provide an additive cellular effect.9 mental signals and are controlled by a circadian timing
mechanism.10 Light is the major environmental cue adjust-
ing the endogenous clock. The retinohypothalamic tract
CONTROL OF HORMONE SECRETION entrains circadian pulse generators situated within hypo-
thalamic suprachiasmatic nuclei. These signals subserve
Anatomically distinct endocrine glands are composed of timing mechanisms for the sleep-wake cycle and determine
highly differentiated cells that synthesize, store, and secrete patterns of hormone secretion and action. Disturbed circa-
hormones. Circulating hormone concentrations are a func- dian timing results in hormonal dysfunction and may also
tion of glandular secretory patterns and hormone clearance be reflective of entrainment or pulse generator lesions. For
rates. Hormone secretion is tightly regulated to attain cir- example, adult GH deficiency due to a damaged hypothala-
culating levels that are most conducive to elicit the appro- mus or pituitary is associated with elevations in integrated
priate target tissue response. For example, longitudinal 24-hour leptin concentrations, decreased leptin pulsatility,
bone growth is initiated and maintained by exquisitely and yet preserved circadian rhythm of leptin. GH replace-
regulated levels of circulating GH, yet mild GH hypersecre- ment restores leptin pulsatility, followed by loss of body
tion results in gigantism, and GH deficiency causes growth fat mass.11 Sleep is also an important cue regulating
retardation. Ambient circulating hormone concentrations hormone pulsatility. About 70% of overall GH secretion
are not uniform, and secretion patterns determine appro- occurs during slow-wave sleep, and increasing age is associ-
priate physiologic function. Thus, insulin secretion occurs ated with declining slow-wave sleep and concomitant
in short pulses elicited by nutrient and other signals; decline in GH and elevation of cortisol secretion.12
gonadotropin secretion is episodic, determined by a hypo- Most pituitary hormones are secreted in a circadian (day-
thalamic pulse generator; and PRL secretion appears to be night) rhythm, best exemplified by ACTH peaks before 9
relatively continuous, with secretory peaks elicited during AM, whereas ovarian steroids follow a 28-day menstrual
suckling. rhythm. Disrupted episodic rhythms are often a hallmark
8 SECTION I  Hormones and Hormone Action

External/internal
environmental
signals

Central nervous
system

Electric or chemical
transmission

Hypothalamus Axonal transport

Releasing hormones (ng) Oxytocin, vasopressin

Adenohypophysis Neurohypophysis

Long Short Release


Anterior pituitary
feedback feedback
hormones (µg)
loop loop

Uterine
Fast Water
Target glands contraction
feedback balance
Lactation
loop (vasopressin)
(oxytocin)

Ultimate hormone (µg-mg)

Hormonal response

Figure 1-3 Peripheral feedback mechanism and a million-fold amplifying cascade of hormonal signals. Environmental signals are transmitted to the central nervous system,
which innervates the hypothalamus, which responds by secreting nanogram amounts of a specific hormone. Releasing hormones are transported down a closed portal system,
pass the blood-brain barrier at either end through fenestrations, and bind to specific anterior pituitary cell membrane receptors to elicit secretion of micrograms of specific
anterior pituitary hormones. These hormones enter the venous circulation through fenestrated local capillaries, bind to specific target gland receptors, trigger release of micro-
grams to milligrams of daily hormone amounts, and elicit responses by binding to receptors in distal target tissues. Peripheral hormone receptors enable widespread cell signaling
by a single initiating environmental signal, thus facilitating intimate homeostatic association with the external environment. Arrows with a large dot at their origin indicate a
secretory process. (From Normal AW, Litwack G. Hormones, 2nd ed. New York: Academic Press; 1997:14.)

of endocrine dysfunction. Thus, loss of circadian ACTH tions are tightly regulated by the effects they elicit. Thus,
secretion with high midnight cortisol levels is a feature of PTH secretion is induced when serum calcium levels fall,
Cushing disease. and the signal for sustained PTH secretion is abrogated by
Hormone secretion is induced by multiple specific bio- rising calcium levels.
chemical and neural signals. Integration of these stimuli Several tiers of control subserve the ultimate net glan-
results in the net temporal and quantitative secretion of dular secretion. First, central nervous system signals includ-
the hormone (Fig. 1-3). Thus, signals elicited by hypotha- ing stress, afferent stimuli, and neuropeptides signal the
lamic hormones (growth hormone–releasing hormone synthesis and secretion of hypothalamic hormones and
[GHRH], SRIF), peripheral hormones (IGF-1, sex steroids, neuropeptides (Fig. 1-4). Four hypothalamic-releasing hor-
thyroid hormone), nutrients, adrenergic pathways, stress, mones (GHRH, corticotropin-releasing hormone [CRH],
and other neuropeptides all converge on the somatotroph thyrotropin-releasing hormone [TRH], and gonadotropin-
cell, resulting in the ultimate pattern and quantity of GH releasing hormone [GnRH]) traverse the hypothalamic
secretion. Networks of reciprocal interactions allow for portal vessels and impinge upon their respective trans-
dynamic adaptation and shifts in environmental signals. membrane trophic hormone-secreting cell receptors.
These regulatory systems embrace the hypothalamic pitu- These distinct cells express GH, ACTH, TSH, and gonado-
itary and target endocrine glands, as well as the adipocytes tropins. In contrast, hypothalamic somatostatin and dopa-
and lymphocytes. Peripheral inflammation and stress elicit mine suppress GH, PRL, and TSH secretion. Trophic
cytokine signals that interface with the neuroendocrine hormones also maintain the structural-functional integrity
system, resulting in hypothalamic-pituitary axis activation. of endocrine organs, including the thyroid and adrenal
The parathyroid and pancreatic secreting cells are less glands and the gonads. Target hormones, in turn, serve as
tightly controlled by the hypothalamus, but their func- powerful negative feedback regulators of their respective
CHAPTER 1  Principles of Endocrinology 9

CNS Inputs or hormone-binding proteins. Alternatively, peripheral


hormone receptor function can be assessed. When a feed-
back loop exists between the hypothalamic-pituitary axis
and a target gland, the circulating level of the pituitary
Tier I
Hypothalamus trophic hormone, such as TSH or ACTH, is typically an
Hypothalamic
hormones exquisitely sensitive index of deficient or excessive func-
tion of the thyroid or the adrenal cortex, respectively.
Meaningful strategies for timing hormonal measurements
vary from system to system. In some cases, circulating
Tier II hormone concentrations can be measured in randomly
Pituitary Paracrine cytokines collected serum samples. This measurement, when stan-
and growth factors dardized for fasting, environmental stress, age, and gender,
is reflective of true hormone concentrations only when
Pituitary trophic levels do not fluctuate appreciably. For example, thyroid
hormone hormone, PRL, and IGF-1 levels can be accurately assessed
Tier III in fasting morning serum samples. On the other hand,
Target gland Peripheral when hormone secretion is clearly episodic, timed samples
hormones may be required over a defined time course to reflect
Figure 1-4 Model for regulation of anterior pituitary hormone secretion by three hormone bioavailability. Thus, early morning and late
tiers of control. Hypothalamic hormones impinge directly on their respective target evening cortisol measurements are most appropriate.
cells. Intrapituitary cytokines and growth factors regulate trophic cell function by Twenty-four–hour sampling for GH measurements, with
paracrine (and autocrine) control. Peripheral hormones exert negative feedback inhibi- samples collected every 2, 10, or 20 minutes, is expensive
tion of respective pituitary trophic hormone synthesis and secretion. CNS, central and cumbersome, yet may yield valuable diagnostic
nervous system. (From Ray D, Melmed S. Pituitary cytokine and growth factor expres- information. Random sampling may also reflect secretion
sion and action. Endocrin Rev. 1997;18:206-228.) peaks or nadirs, thus confounding adequate interpretation
of results.
trophic hormone; they often also suppress secretion of In general, confirmation of failed glandular function is
hypothalamic-releasing hormones. In certain circum- made by attempting to evoke hormone secretion by recog-
stances (e.g., during puberty), peripheral sex steroids may nized stimuli. Thus, testing of pituitary hormone reserve
positively induce the hypothalamic-pituitary-target gland may be accomplished by injecting appropriate hypotha-
axis. Thus, LH induces ovarian estrogen secretion, which lamic releasing hormones. Injection of trophic hormones,
feeds back positively to induce further LH release. Pituitary including TSH and ACTH, evokes specific target gland
hormones themselves, in a short feedback loop, may also hormone secretion. Pharmacologic stimuli (e.g., metoclo-
regulate their own respective hypothalamic-controlling pramide for induction of PRL secretion) may also be useful
hormone. Hypothalamic-releasing hormones are secreted tests of hormone reserve. In contrast, hormone hypersecre-
in nanogram amounts and have short half-lives of a few tion can be diagnosed by suppressing glandular function.
minutes. Anterior pituitary hormones are produced in Thus, failure to appropriately suppress GH levels after a
microgram amounts and have longer half-lives, but periph- standardized glucose load implies inappropriate GH hyper-
eral hormones can be produced in up to milligram amounts secretion. The failure to suppress insulin secretion in
daily, with much longer half-lives. response to hypoglycemia indicates inappropriate hyperse-
A further level of secretion control occurs within the cretion of insulin and should prompt a search for the cause,
gland itself. Thus, intraglandular paracrine or autocrine such as an insulin-secreting tumor.
growth peptides serve to autoregulate pituitary hormone Radioimmunoassays use highly specific antibodies that
secretion, as exemplified by epidermal growth factor (EGF) uniquely recognize the hormone, or a hormone fragment,
control of PRL or IGF-1 control of GH secretion. Molecules to quantify hormone levels. Enzyme-linked immunosor-
within the endocrine cell may also subserve an intracel- bent assays (ELISAs) employ enzymes instead of radioactive
lular feedback loop. Thus, corticotrope SOCS-3 induction hormone markers, and enzyme activity is reflective of hor-
by gp130-linked cytokines serves to abrogate the ligand- mone concentration. This sensitive technique has allowed
induced JAK-STAT cascade and block pro-opiomelanocortin ultrasensitive measurements of physiologic hormone con-
(POMC) transcription and ACTH secretion. This rapid centrations. Hormone-specific receptors may be employed
on-off regulation of ACTH secretion provides a plastic in place of the antibody in a radioreceptor assay.
endocrine response to changes in environmental signaling
and serves to maintain homeostatic integrity.13
In addition to the central nervous system–neuroendocrine
interface mediated by hypothalamic chemical signal trans- ENDOCRINE DISEASES
duction, the central nervous system directly controls
several hormonal secretory processes. Posterior pituitary Endocrine diseases fall into four broad categories: (1)
hormone secretion occurs as direct efferent neural exten- hormone overproduction, (2) hormone underproduction,
sions. Postganglionic sympathetic nerves also regulate (3) altered tissue responses to hormones, and (4) tumors of
rapid changes in renin, insulin, and glucagon secretion, endocrine glands. An additional fifth category is so far
and preganglionic sympathetic nerves signal to adrenal exemplified by one kind of hypothyroidism in which over-
medullary cells eliciting adrenaline release. expression of a hormone-inactivating enzyme in a tumor
leads to thyroid hormone deficiency.

HORMONE MEASUREMENT Hormone Overproduction


Occasionally, hormones are secreted in increased amounts
Endocrine function can be assessed by measuring levels of because of genetic abnormalities that cause abnormal regu-
basal circulating hormone, evoked or suppressed hormone, lation of hormone synthesis or release. For example, in
10 SECTION I  Hormones and Hormone Action

glucocorticoid-remediable hyperaldosteronism, an abnor- vating mutations in the Gsα protein can cause precocious
mal chromosomal crossing over event puts the aldosterone puberty, hyperthyroidism, and acromegaly in McCune-
synthetase gene under the control of the ACTH-regulated Albright syndrome.
11β-hydroxylase gene. More often, diseases of hormone
overproduction are associated with an increase in the total
number of hormone-producing cells. For example, the
Tumors of Endocrine Glands
hyperthyroidism of Graves disease, in which antibodies Tumors of endocrine glands, as noted previously, often
mimic TSH and activate the TSH receptors on thyroid cells, result in hormone overproduction. Some tumors of endo-
is associated with dramatic increase in thyroid cell prolif- crine glands produce little if any hormone but cause disease
eration, as well as with increased synthesis and release of by causing local compressive symptoms or by metastatic
thyroid hormone from each thyroid cell. In this example, spread. Examples include so-called nonfunctioning pitu-
the increase in thyroid cell number represents a polyclonal itary tumors, which are usually benign but can cause a
expansion of thyroid cells, in which large numbers of variety of symptoms due to compression on adjacent struc-
thyroid cells proliferate in response to an abnormal stimu- tures, and thyroid cancer, which can spread throughout
lus. However, most endocrine tumors are not polyclonal the body without causing hyperthyroidism.
expansions, but instead represent monoclonal expansions
of one mutated cell. Pituitary and parathyroid tumors, for Excessive Hormone Inactivation
example, are usually monoclonal expansions in which
somatic mutations in multiple tumor suppressor genes and
or Destruction
proto-oncogenes occur. These mutations lead to an increase Although most enzymes important for endocrine systems
in proliferation or survival of the mutant cells. Sometimes, activate a prohormone or precursor protein, there are also
this proliferation is associated with abnormal secretion of those whose function is to inactivate the hormone in a
hormone from each tumor cell as well. For example, physiologically regulated fashion. An example is the type
mutant Gsα proteins in somatotrophs can lead to both 3 iodothyronine deiodinase (D3), which inactivates T3 and
increased cellular proliferation and increased secretion of T4 by removing an inner ring iodine atom from the iodo-
GH from each tumor cell. thyronine, blocking its nuclear receptor binding. Large
infantile hepatic hemangiomas express high D3 levels,
causing “consumptive hypothyroidism,” so named because
Hormone Underproduction thyroid hormone is inactivated at a more rapid rate than
Underproduction of hormone can result from a wide it can be produced.14,15 Furthermore, D3 may also be
variety of processes, ranging from surgical removal of para- induced in other tumors by tyrosine kinase inhibitors. In
thyroid glands during neck surgery, to tuberculous destruc- theory, accelerated destruction of other hormones could
tion of adrenal glands, or to iron deposition in β cells of occur from similar processes, but no examples have been
islets in hemochromatosis. A frequent cause of destruction reported to date.
of hormone-producing cells is autoimmunity. Autoim-
mune destruction of β cells in type 1 diabetes mellitus or
of thyroid cells in Hashimoto thyroiditis are two of the
most common disorders treated by endocrinologists. More DIAGNOSTIC AND THERAPEUTIC USES
uncommonly, a host of genetic abnormalities can also lead OF HORMONES
to decreased hormone production. These disorders can
result from abnormal development of hormone-producing In general, hormones are employed pharmacologically for
cells (e.g., hypogonadotropic hypogonadism caused by both their replacement or suppressive effects. Hormones
KAL gene mutations), from abnormal synthesis of hor- may also be used for diagnostic stimulatory effects (e.g.,
mones (e.g., deletion of the GH gene), or from abnormal hypothalamic hormones) to evoke target organ responses,
regulation of hormone secretion (e.g., the hypoparathy- or to diagnose endocrine hyperfunction by suppressing
roidism associated with activating mutations of the para- hormone hypersecretion (e.g., T3). Ablation of endocrine
thyroid cell’s calcium-sensing receptor). gland function due to genetic or acquired causes can be
restored by hormone replacement therapy. In general,
steroid and thyroid hormones are replaced orally, whereas
Altered Tissue Responses peptide hormones (e.g., insulin, GH) require injection.
Resistance to hormones can be caused by a variety of Gastrointestinal absorption and first-pass kinetics deter-
genetic disorders. Examples include mutations in the GH mine oral hormone dosage and availability. Physiologic
receptor in Laron dwarfism and mutations in the Gsα replacement can achieve both appropriate hormone levels
gene in the hypoparathyroidism of pseudohypoparathy- (e.g., thyroid) as well as approximate hormone secretory
roidism type 1A. The insulin resistance in muscle and patterns (e.g., GnRH delivered intermittently via a pump).
liver central to the cause of type 2 diabetes mellitus is Hormones can also be used to treat diseases associated
complex in origin, resulting from inherited variations in with glandular hyperfunction. Long-acting depot prepara-
many genes, as well as from theoretically reversible physi- tions of somatostatin analogues suppress GH hypersecre-
ologic stresses. Type 2 diabetes is also an example of a tion in acromegaly or 5-hydroxyindoleacetic acid (5-HIAA)
disease in which end-organ insensitivity is worsened by hypersecretion in carcinoid syndrome. Estrogen receptor
signals from other organs, in this case by signals originat- antagonists (e.g., tamoxifen) are useful for some patients
ing in fat cells. In other cases, the target organ of hormone with breast cancer, and GnRH analogues may downregu-
action is more directly abnormal, as in the PTH resistance late the gonadotropin axis and benefit patients with pros-
of renal failure. tate cancer.
Increased end-organ function can be caused by muta- Novel formulations of receptor-specific hormone ligands
tions in signal reception and propagation. For example, are now being clinically developed (e.g., estrogen agonists/
activating mutations in TSH, LH, and PTH receptors can antagonists, somatostatin receptor subtype ligands), result-
cause increased activity of thyroid cells, Leydig cells, and ing in more selective therapeutic targeting. Modes of
osteoblasts, even in the absence of ligand. Similarly, acti- hormone injection (e.g., for PTH) may also determine
CHAPTER 1  Principles of Endocrinology 11

therapeutic specificity and efficacy. Improved hormone when many children with that diagnosis achieve adult-
delivery systems, including computerized minipumps, hood means either that we have little understanding of the
intranasal sprays (e.g., for desmopressin [DDAVP]), pulmo- etiology/pathogenesis of that deficiency or that our diag-
nary inhalers, depot intramuscular injections, and oral nostic tools today have many false-positive results.
peptide formulations, will also enhance patient compliance Although endocrinologists pride themselves with having
and improve ease of administration. Insulin delivered logical treatments for many diseases, these treatments
by inhalation has already been approved for use, and seldom address their underlying causes. We have no satis-
inhaled GH and oral octreotide are under investigation. factory tools for preventing autoimmune endocrine defi-
Cell-based therapies using the reprogramming of human ciencies or for preventing the benign tumors that underlie
cells to perform differentiated functions, either through many diseases characterized by hormone excess. Treat-
differentiation of induced pluripotent stem cells or directed ments for diseases such as type 1 diabetes, although highly
differentiation of one somatic cell type into another, are effective, are still very obtrusive in the lives of patients with
under active investigation.16 Novel technologies offer this disease.
promise of marked prolongation in the half-life of peptide Although the primary rationale for this new edition is
hormones, thereby requiring infrequent administration. to communicate the major advances that have been made
For example, a once weekly preparation of exenatide, a in our field over the past 5 years, large gaps in our knowl-
glucagon-like peptide-1 (GLP-1) analogue currently used in edge about endocrinology remain. While this realization is
the treatment of type 2 diabetes, is currently undergoing sobering, we hope that it will be viewed by our readers as
clinical trials. an exciting challenge for the future. That is the spirit
Tremendous progress has been made in the therapeutic underlying this text.
use of hormones. Although the delivery of insulin still
requires frequent administration by injection and close
monitoring by the patient, the purity of the insulin prepa- REFERENCES
rations has been vastly improved, thereby almost entirely 1. Liu PT, Stenger S, Li H, et  al. Toll-like receptor triggering of a vita-
eliminating the occurrence of uncomfortable local reac- min D-mediated human antimicrobial response. Science. 2006;311:
1770-1773.
tions at the injection site. Preparations with differing 2. Chawla A, Repa JJ, Evans RM, et al. Nuclear receptors and lipid physiol-
pharmacokinetics allow the normal physiology of insulin ogy: opening the X-files. Science. 2001;294:1866-1870.
secretion to be more closely mimicked. Continuous admin- 3. Mendel CM, Weisiger RA, Jones AL, Cavalieri RR. Thyroid hormone-
istration via subcutaneous pump infusion greatly enhances binding proteins in plasma facilitate uniform distribution of thyroxine
within tissues: a perfused rat liver study. Endocrinology. 1987;120(5):
therapeutic effectiveness in carefully selected patients. 1742-1749.
Closed-loop systems, in which the dose of insulin could be 4. Mendel CM. The free hormone hypothesis: physiologically based math-
regularly adjusted depending on blood glucose concentra- ematical model. Endocr Rev. 1989;10(3):232-274.
tions, are being actively studied. The implementation of 5. Safadi FF, Thornton P, Magiera H, et al. Osteopathy and resistance to
vitamin D toxicity in mice null for vitamin D binding protein. J Clin
such systems in the future would substantially reduce the Invest. 1999;103:239-251.
burden of this disease. Hormones are biologically powerful 6. Palha JA, Fernandes R, de Escobar GM, et al. Transthyretin regulates
molecules that exert therapeutic benefit and effectively thyroid hormone levels in the choroid plexus, but not in the brain
replace pathologic deficits. They should not be prescribed parenchyma: study in a transthyretin-null mouse model. Endocrinology.
2000;141:3267-3272.
without clear-cut indications and should not be adminis- 7. Mayerl S, Müller J, Bauer R, et al. Transporters MCT8 and OATP1C1
tered without careful evaluation by an appropriately quali- maintain murine brain thyroid hormone homeostasis. J Clin Invest.
fied medical practitioner. 2014;124:1987-1999.
8. Hammes A, Andreassen TK, Spoelgen R, et al. Role of endocytosis in
cellular uptake of sex steroids. Cell. 2005;122:751-762.
9. Rocheville M, Lange DC, Kumar U, et al. Receptors for dopamine and
WHAT WE DON’T KNOW (YET) somatostatin: formation of hetero-oligomers with enhanced functional
activity. Science. 2000;288:154-157.
An introduction to the principles underlying endocrinol- 10. Moore RY. Circadian rhythms: basic neurobiology and clinical applica-
tions. Annu Rev Med. 1997;48:253-266.
ogy should end by emphasizing the rapidly changing 11. Aftab MA, Guzder R, Wallace AM, et al. Circadian and ultradian rhythm
dynamics of discovery in this field and attempting to and leptin pulsatility in adult GH deficiency: effects of GH replacement.
foresee what remains to be discovered. New hormones are J Clin Endocrinol Metab. 2001;86:3499-3506.
continually being discovered, from the recent focus on 12. Cauter EV, Leproult R, Plat L. Age-related changes in slow wave sleep
and REM sleep and relationship with growth hormone and cortisol
major regulators of metabolism and phosphate homeosta- levels in healthy men. JAMA. 2000;284:861-868.
sis (FGF19, -21, and -23) to the continued quest to identify 13. Melmed S. The immuno-neuroendocrine interface. J Clin Invest. 2001;
ligands for “orphan” nuclear and G protein–coupled recep- 108:1563-1566.
tors.17 Presumably, other equally important hormones 14. Huang SA, Tu HM, Harney JW, et al. Severe hypothyroidism caused by
type 3 iodothyronine deiodinase (D3) in infantile hemangiomas. N Engl
remain to be discovered. The observation that nuclear J Med. 2000;343:185-189.
receptors, like most transcription factors, bind to thou- 15. Bianco AC, Salvatore D, Gereben B, et al. Biochemistry, cellular and
sands of specific sites within the cell’s nucleus stresses how molecular biology, and physiological roles of the iodothyronine seleno-
little we understand about hormone action. Many of our deiodinases. Endocr Rev. 2002;23:38-89.
16. Pagliuca FW, Melton DA. How to make a functional β-cell. Development.
diagnostic tests are severely limited by both technology as 2013;140:2472-2483.
well as our ability to foresee novel diagnostic targets. For 17. Evans RM, Mangelsdorf DJ. Nuclear receptors. RXR, and the Big Bang.
example, the “disappearance” of isolated GH deficiency Cell. 2014;157:255-266.

Vous aimerez peut-être aussi