You are on page 1of 10

The Evolutionary Perspective,  3

Endocrine Glands,  5
Transport of Hormones in Blood,  6
Target Cells as Active Participants,  7
Control of Hormone Secretion,  8
Hormone Measurement,  10
Endocrine Diseases,  11
Therapeutic Strategies,  12

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

Roughly 100 years ago, Starling coined the term hormone diverse approaches used by clinicians, physiologists, bio-
to describe secretin, a substance secreted by the small intes- chemists, cell biologists, and geneticists to understand the
tine into the bloodstream to stimulate pancreatic secretion. endocrine system.
In his Croonian Lectures, Starling considered the endo-
crine and nervous systems as two distinct mechanisms for
coordination and control of organ function. Thus, endo- THE EVOLUTIONARY PERSPECTIVE
crinology found its first home in the discipline of mam-
malian physiology. Hormones can be defined as chemical signals secreted into
Work over the next several decades by biochemists, the bloodstream that act on distant tissues, usually in a
physiologists, and clinical investigators led to the charac- regulatory fashion. Hormonal signaling represents a special
terization of many hormones secreted into the bloodstream case of the more general process of signaling between cells.
from discrete glands or other organs. Investigations showed Even unicellular organisms, such as baker’s yeast, Saccharo-
that diseases such as hypothyroidism and diabetes could myces cerevisiae, secrete short peptide mating factors that
be treated successfully, for the first time, by replacing spe- act on receptors of other yeast cells to trigger mating
cific hormones. These initial triumphs formed the founda- between the two cells. These receptors resemble the ubiq-
tion of the clinical specialty of endocrinology. uitous family of mammalian seven-transmembrane span-
Advances in cell biology, molecular biology, and genet- ning receptors that respond to ligands as diverse as photons
ics over the ensuing years began to explain the mecha- and glycoprotein hormones. Because these yeast receptors
nisms of endocrine diseases and of hormone secretion and trigger activation of heterotrimeric G proteins just as mam-
action. Even though these advances have embedded endo- malian receptors do, this conserved signaling pathway
crinology in the framework of molecular cell biology, they must have been present in the common ancestor of yeast
have not changed the essential subject of endocrinology— and humans.
the signaling mechanisms that coordinate and control the Signals from one cell to adjacent cells—so-called para-
functions of multiple organs and processes. Herein we crine signals—often trigger cellular responses that use the
survey the general themes and principles that underpin the same molecular pathways used by hormonal signals. For
3
4   Principles of Endocrinology

example, the sevenless receptor controls the differentiation speculate that the hormonal actions of vitamin D might
of retinal cells in the Drosophila eye by responding to a have evolved well after the paracrine vitamin D apparatus
membrane-anchored signal from an adjacent cell. Seven- provided the raw materials for the hormonal system.
less is a membrane-spanning receptor with an intracellular Target cells respond similarly to signals that reach them
tyrosine kinase domain that signals in a way that closely from the bloodstream (hormones) or from the cell next
resembles the signaling by hormone receptors such as the door (paracrine factors); the cellular response machinery
insulin receptor tyrosine kinase. does not distinguish the sites of origin of signals. The
Because paracrine factors and hormones can share sig- shared final common pathways used by hormonal and
naling machinery, it is not surprising that hormones can, paracrine signals should not, however, obscure important
in some settings, act as paracrine factors. Testosterone, for differences between hormonal and paracrine signaling
example, is secreted into the bloodstream but also acts systems (Fig.1-1). Paracrine signals do not travel very far;
locally in the testes to control spermatogenesis. Insulin-like consequently, the specific site of origin of a paracrine factor
growth factor 1 (IGF1) is a polypeptide hormone that is determines where it will act and provides specificity to that
secreted into the bloodstream from the liver and other action. When the paracrine factor bone morphogenic
tissues, but it is also a paracrine factor made locally in most protein 4 (BMP4) is secreted by cells in the developing
tissues to control cell proliferation. kidney, BMP4 regulates the differentiation of renal cells;
Furthermore, one receptor can mediate actions of a when the same factor is secreted by cells in bone, it regu-
hormone and a paracrine factor, such as parathyroid lates bone formation. Thus, the site of origin of BMP4
hormone (PTH) and parathyroid hormone–related protein. determines its physiologic role. In contrast, because hor-
In some cases, the paracrine actions of “hormones” have mones are secreted into the bloodstream, their sites of
functions quite unrelated to hormonal functions. For origin are often divorced from their functions. There is
example, macrophages synthesize the active form of nothing about thyroid hormone function, for example,
vitamin D, 1,25-dihydroxyvitamin D3 or calcitriol, which that requires that the thyroid gland be in the neck.
can then bind to vitamin D receptors in the same cells and Because the specificity of paracrine factor action is so
stimulate production of antimicrobial peptides.1 The dependent on its precise site of origin, elaborate mecha-
vitamin D 1α-hydroxylase responsible for activating 25- nisms have evolved to regulate and constrain the diffusion
hydroxyvitamin D (calcidiol) is synthesized in multiple of paracrine factors. Paracrine factors of the hedgehog
tissues in which it has functions not apparently related to family, for example, are covalently bound to cholesterol
the calcium homeostatic actions of calcitriol. One can to constrain the diffusion of these molecules in the

Regulation of signaling: endocrine


Source: gland Distribution: blood stream 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
• Proteases Figure 1-1 Comparison of determinants of endocrine and
• Matrix components paracrine signaling.
Principles of Endocrinology   5

extracellular milieu. Most paracrine factors interact with which no clear ligand or function is known. The analyses
binding proteins that block their action and control their of these “orphan” receptors have succeeded in broadening
diffusion. For example, chordin, noggin, and many other the current understanding of hormonal signaling. For
distinct proteins bind to various members of the BMP example, the orphan liver X receptor, LXR, was found
family to regulate their action. Proteases such as tolloid during searches for unknown nuclear receptors. Subse-
then destroy the binding proteins at specific sites to liber- quent experiments showed that oxygenated derivatives of
ate BMPs so that they can act on appropriate target cells. cholesterol are the ligands for LXR, which regulates genes
Hormones have rather different constraints. Because involved in cholesterol and fatty acid metabolism.2 LXR
they diffuse throughout the body, they must be synthesized and many other examples raise the question of what
in enormous amounts relative to the amounts of paracrine constitutes a hormone.
factors needed at specific locations. This synthesis usually The classic view of hormones is that they are synthe-
occurs in specialized cells designed for that specific purpose. sized in discrete glands and have no function other than
Hormones must then be able to travel in the bloodstream activating receptors on cell membranes or in the nucleus.
and diffuse in effective concentrations into tissues. Lipo- In contrast, cholesterol, which is converted in cells to oxy-
philic hormones, for example, bind to soluble proteins that genated derivatives that activate the LXR receptor, uses a
allow them to travel in the aqueous medium of blood at hormonal strategy to regulate its own metabolism. Other
relatively high concentrations. The ability of hormones to orphan nuclear receptors similarly respond to ligands such
diffuse through the extracellular space means that the local as bile acids and fatty acids. These “hormones” have impor-
concentration of a hormone at target sites will rapidly tant metabolic roles quite separate from their signaling
decrease when glandular secretion of the hormone stops. properties, although the hormone-like signaling serves to
Because hormones diffuse throughout extracellular fluid allow regulation of the metabolic function. The calcium-
quickly, hormonal metabolism can occur in specialized sensing receptor is an example from the G protein–linked
organs (e.g., liver, kidney) in a manner that determines the receptor family that responds to a nonclassic ligand, ionic
effective hormone concentration in other tissues. calcium. Calcium is released into the bloodstream from
In summary, paracrine factors and hormones use several bone, kidney, and intestine and acts on the calcium-sensing
distinct strategies to control their biosynthesis, sites of receptors on parathyroid cells, renal tubular cells, and
action, transport, and metabolism. These differing strate- other cells to coordinate cellular responses to calcium.
gies probably explain partly why a hormone such as IGF1, Therefore, many important metabolic factors have taken
unlike its close relative, insulin, has multiple binding on hormonal properties as part of a regulatory strategy.
proteins that control its action in tissues. IGF1 exhibits a
double life—it is both a hormone and a paracrine factor.
Presumably, the local actions of IGF1 mandate an elaborate ENDOCRINE GLANDS
binding protein apparatus to enable appropriate hormone
signaling. Hormone formation may occur either in localized collec-
All of the major hormonal signaling programs—G tions of specific cells, the endocrine glands, or in cells that
protein–coupled receptors, tyrosine kinase receptors, have additional roles. Many protein hormones, such as
serine/threonine kinase receptors, ion channels, cytokine growth hormone (GH), PTH, prolactin, insulin, and gluca-
receptors, and nuclear receptors—are also used by para- gon, are produced in dedicated cells by standard protein
crine factors. In contrast, several paracrine signaling pro- synthetic mechanisms common to all cells. These secretory
grams appear to be used only by paracrine factors and not cells usually contain specialized secretory granules designed
by hormones. For example, Notch receptors respond to to store large amounts of hormone and to release the
membrane-based ligands to control cell fate, but no blood- hormone in response to specific signals. Formation of small
borne ligands are known to use Notch-type signaling. hormone molecules begins with commonly found precur-
Perhaps the intracellular strategy used by Notch, which sors, usually located in specific glands such as the adrenals,
involves cleavage of the receptor and subsequent nuclear gonads, or thyroid. In the case of the steroid hormones,
actions of the receptor’s cytoplasmic portion, is too inflex- the precursor is cholesterol, which is modified by various
ible to serve the purposes of hormones. hydroxylations, methylations, and demethylations to form
The analyses of the complete genomes of multiple bac- glucocorticoids, androgens, estrogens, and their biologi-
terial species, the yeast S. cerevisiae, the fruit fly Drosophila cally 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 earlier, S. cerevisiae expenditure, is formed in adipocytes, providing a
uses G protein–linked receptors; this organism, however, specific signal that reflects the nutritional state to the
lacks tyrosine kinase receptors and nuclear receptors that 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- In the unique enteroendocrine hormonal system, peptide
tion in the number of genes committed to each pathway. hormones that regulate metabolic and other responses to
For example, the Drosophila genome encodes 20 nuclear oral nutrients are produced and secreted by specialized
receptors, the C. elegans genome 270, and the human endocrine cells scattered throughout the intestinal
genome (tentatively) more than 50. These patterns suggest epithelium.
that ancient multicellular animals must have already estab- Thyroid hormone synthesis occurs by means of a unique
lished the signaling systems that are the foundation of the pathway. The thyroid cell synthesizes a 660,000-kd homodi-
endocrine system as we know it in mammals. mer, thyroglobulin, which is then iodinated at specific
Even before the sequencing of the human genome was iodotyrosines. Certain of these “couple” to form the iodo-
accomplished, sequence analyses had made clear that thyronine molecule within thyroglobulin, which is then
many receptor genes are found in mammalian genomes for stored in the lumen of the thyroid follicle. For this to occur,
6   Principles of Endocrinology

the thyroid cell must concentrate the trace quantities of programmed increases and decreases in hormone secretion
iodide from the blood and oxidize it via a specific peroxi- or activation also occur through neuroendocrine pathways.
dase. Release of thyroxine (T4) from the thyroglobulin Examples include the circadian variation in secretion of
requires its phagocytosis and cathepsin-catalyzed digestion ACTH that directs the synthesis and release of cortisol. The
by the same cells. monthly menstrual cycle exemplifies a system with much
Hormones are synthesized in response to biochemical longer periodicity that requires a complex synergism
signals generated by various modulating systems. Many of between central and peripheral axes of the endocrine
these systems are specific to the effects of the hormone glands. Disruption of hormonal homeostasis due to
product. For example, PTH synthesis is regulated by the glandular or central regulatory system dysfunction has
concentration of ionized calcium, and insulin synthesis is both clinical and laboratory consequences. Recognition
regulated by the concentration of glucose. For others, such and correction of these effects are the essence of clinical
as gonadal, adrenal, and thyroid hormones, control of 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
concentration and secrete tropic hormones, which activate IN BLOOD
specific pathways for hormone synthesis and release.
Typical examples are luteinizing hormone (LH), follicle- Protein hormones and some small molecules, such as the
stimulating hormone (FSH), thyroid-stimulating hormone catecholamines, are water soluble and readily transported
(TSH), and adrenocorticotropic hormone (ACTH). via the circulatory system. Others, such as the steroid and
These trophic hormones increase rates of hormone syn- thyroid hormones, are almost insoluble in water, and their
thesis and secretion, and they may induce target cell divi- distribution presents special problems. Such molecules are
sion, resulting in enlargement of the various target glands. bound to 50- to 60-kd carrier plasma glycoproteins such as
For example, in hypothyroid individuals living in iodine- thyroxine-binding globulin (TBG), sex hormone–binding
deficient areas of the world, TSH secretion causes a marked globulin (SHBG), and corticosteroid-binding globulin, as
hyperplasia of thyroid cells. In such regions, the thyroid well as to albumin. The ligand-protein complexes serve as
gland may be 20 to 50 times its normal size. Adrenal hyper- reservoirs of these hormones, ensure ubiquitous distribu-
plasia occurs in patients with genetic deficiencies in corti- tion of their water-insoluble ligands, and protect the small
sol formation. Hypertrophy and hyperplasia of parathyroid molecules from rapid inactivation or excretion in the urine
cells, in this case initiated by an intrinsic response to the or bile. Without these proteins, it is unlikely that hydro-
stress of hypocalcemia, occurs in patients with renal insuf- phobic molecules would be transported much beyond the
ficiency or calcium malabsorption. veins draining the glands in which they are formed.
Hormones may be fully active when they are released The protein-bound hormones exist in rapid equilibrium
into the bloodstream (e.g., GH, insulin), or they may with the often minute quantities of hormone in the
require activation in specific cells to produce their biologic aqueous plasma. It is this “free” fraction of the circulating
effects. These activation steps are often highly regulated. hormone that is taken up by the cell. For example, if tracer
For example, the T4 released from the thyroid cell is a pro- thyroid hormone is injected into the portal vein in a
hormone that must undergo a specific deiodination to protein-free solution, it becomes bound to hepatocytes at
form the active 3,5,3′-triiodothyronine (T3). This deiodin- the periphery of the hepatic sinusoid. When the same
ation reaction can occur in target tissues (e.g., in the central experiment is repeated with a protein-containing solution,
nervous system); in the thyrotrophs, where T3 provides there is a uniform distribution of tracer hormone through-
feedback regulation of TSH production; or in hepatic and out the hepatic lobule.3
renal cells, from which it is released into the circulation for Despite the very high affinity of some of the binding
uptake by all tissues. A similar postsecretory activation proteins for their ligands, a specific protein may not be
step, catalyzed by a 5αα-reductase, causes tissue-specific essential for hormone distribution. For example, in humans
activation of testosterone to dihydrotestosterone in target with a congenital deficiency of TBG, other proteins, namely
tissues including the male urogenital tract and genital skin, transthyretin (TTR) and albumin, subsume its role. Because
as well as in liver. Vitamin D undergoes hydroxylation at the affinity of these secondary thyroid hormone transport
the 25 position in the liver and at the 1 position in the proteins is several orders of magnitude lower than that of
kidney. Both hydroxylations must occur to produce the TBG, it is possible for the hypothalamic-pituitary feedback
active hormone, calcitriol. The activity of 1α-hydroxylase, system to maintain free thyroid hormone in the normal
but not that of 25-hydroxylase, is stimulated by PTH and range at a much lower total hormone concentration. The
reduced plasma phosphate but inhibited by calcium, fact that the level of “free” hormone concentration is
calcitriol, and fibroblast growth factor 23 (FGF23). normal in subjects with TBG deficiency indicates that it is
Hormones are synthesized as required on a daily, hourly, this free moiety that is defended by the hypothalamic-
or minute-to-minute basis with minimal storage, but there pituitary axis and is the active hormone.4
are significant exceptions. One is the thyroid gland, which The availability of gene targeting techniques has allowed
contains enough stored hormone to last for about 2 specific tests of the physiologic roles of several hormone-
months. This permits a constant supply despite significant binding proteins. For example, mice with targeted inactiva-
variations in the availability of iodine. However, if iodine tion of the vitamin D–binding protein (DBP) have been
deficiency is prolonged, the normal reservoirs of T4 can be generated.5 Although the absence of DBP markedly reduces
depleted. the circulating concentration of vitamin D, the mice are
The various feedback signaling systems already described otherwise normal. However, they do show enhanced sus-
enable the hormonal homeostasis that is characteristic of ceptibility to a vitamin D–deficient diet because of the
virtually all endocrine systems. Regulation may include the reduced reservoir of this sterol. In addition, the absence of
central nervous system or local signal recognition mecha- DBP markedly reduces the half-life of calcidiol by accelerat-
nisms in the glandular cells, such as the calcium-sensing ing its hepatic uptake, making the mice less susceptible to
receptor of the parathyroid cell. Superimposed, centrally vitamin D intoxication.
Principles of Endocrinology   7

In rodents, TTR carries retinol-binding protein and is it is the hormone bound to SHBG, rather than the “free”
also the principal thyroid hormone–binding protein. This hormone, that is the active moiety that enters cells. It is
protein is synthesized in the liver and in choroid plexus. unclear how generally this apparent exception to the “free
It is the major thyroid hormone–binding protein in the hormone” hypothesis occurs.
cerebrospinal fluid of both rodents and humans and was
previously thought to possibly serve an important role in
thyroid hormone transport into the central nervous system. TARGET CELLS AS ACTIVE
This hypothesis was later disproved by the fact that mice PARTICIPANTS
without TTR have normal concentrations of T4 in the brain
in addition to free T4 in the plasma.6,7 To be sure, the serum Hormones determine cellular target actions by binding
concentrations of vitamin A and total T4 are decreased, but with high specificity to receptor proteins. Whether a
the knockout mice have no signs of vitamin A deficiency peripheral cell is hormonally responsive depends to a large
or hypothyroidism. Such studies suggest that these pro- extent on the presence and function of specific and selec-
teins primarily serve distributive and reservoir functions. tive hormone receptors. Receptor expression determines
Protein hormones and some small ligands (e.g., cate- which cells will respond as well as the nature of the intra-
cholamines) produce their effects by interacting with cell cellular effector pathways activated by the hormone signal.
surface receptors. Others, such as the steroid and thyroid Receptor proteins may be localized to the cell membrane,
hormones, must enter the cell to bind to cytosolic or cytoplasm, or nucleus. Broadly, polypeptide hormone
nuclear receptors. In the past, it was thought that much of receptors are associated with cell membranes, whereas
the transmembrane transport of hormones was passive. steroid hormones selectively bind soluble intracellular pro-
Evidence has now demonstrated that specific transporters teins (Fig. 1-2). However, exceptions do occur. For example,
are involved in cellular uptake of thyroid hormone.8 This epidermal growth factor (EGF) may signal directly to recep-
may be found to be the case for other small ligands as well, tors located within the nucleus.
revealing yet another mechanism for ensuring the distribu- Membrane-associated receptor proteins usually consist
tion of a hormone to its site of action. Studies in mice of extracellular sequences that recognize and bind ligand,
missing megalin, a large cell surface protein in the low- transmembrane anchoring hydrophobic sequences, and
density lipoprotein (LDL) receptor family, have suggested intracellular sequences that initiate intracellular signaling.
that estrogen and testosterone uses megalin to enter certain Intracellular signaling is mediated by covalent modifica-
tissues while still bound to SHBG.9 In this case, therefore, tion and activation of intracellular signaling molecules

Progesterone

O R

O RR O
XTyr

Figure 1-2  Hormonal signaling by cell surface and intracel- TF P TFTyr P


R ss
lular receptors. The receptors for the water-soluble poly- PKA s
peptide hormones, luteinizing hormone (LH), and insulin-like Target gene
growth factor 1 (IGF-1), are integral membrane proteins IGF-1
XTyr P
located at the cell surface. They bind the hormone-utilizing cAMP
extracellular sequences and transduce a signal through the
generation of second messengers: cyclic adenosine mono-
phosphate (cAMP) for the LH receptor and tyrosine-
AC

phosphorylated substrates for the IGF-1 receptor. Although ATP AAAAA


effects on gene expression are indicated, direct effects on G mRNAs
cellular proteins (e.g., ion channels) are also observed. In R
s ss

contrast, the receptor for the lipophilic steroid hormone,


progesterone, resides in the cell nucleus. It binds the LH
hormone and becomes activated and capable of directly
modulating target gene transcription.) AC, Adenylate cyclase; Proteins
G, heterotrimeric G protein; mRNAs, messenger RNAs;
PKA, protein kinase A; R, receptor molecule;TF, transcription
factor; Tyr, tyrosine found in protein X; X, unknown protein
substrate. (Reproduced from Mayo K. Receptors: molecular
mediators of hormone action. In: Conn PM, Melmed S, eds.
Endocrinology: Basic and Clinical Principles. Totowa, NJ: Humana
Press, 1997:11.) Biological responses
8   Principles of Endocrinology

(e.g., STAT proteins) or by generation of small molecule may be induced by activating mutations (e.g., TSH recep-
second messengers (e.g., cyclic adenosine monophosphate) tor) leading to endocrine organ hyperfunction, even in the
through activation of heterotrimeric G proteins. The α-, β-, absence of hormone. Conversely, inactivating receptor
and γ-subunits of these G proteins activate or suppress mutations may lead to endocrine hypofunction (e.g., tes-
effector enzymes and ion channels that generate the second tosterone receptor, vasopressin receptor). These syndromes
messengers. Some of these receptors may in fact exhibit are well characterized and are well described in other chap-
constitutive activity and have been shown to signal in the ters of this text (Fig. 1-3).
absence of added ligand. The functional diversity of receptor signaling also results
Several growth factors and hormone receptors (e.g., for in overlapping or redundant intracellular pathways. For
insulin) behave as intrinsic tyrosine kinases or activate example, both GH and cytokines activate JAK-STAT signal-
intracellular protein tyrosine kinases. Ligand activation ing, whereas the distal effects of these stimuli clearly differ.
may cause receptor dimerization (e.g., GH) or heterodimer- Therefore, despite common signaling pathways, hormones
ization (e.g., interleukin-6), followed by activation of intra- elicit highly specific cellular effects. Tissue or cell-type
cellular phosphorylation cascades. These activated proteins genetic programs or receptor-receptor interactions at the
ultimately determine specific nuclear gene expression. cell surface (e.g., dopamine D2 hetero-oligomerization with
Both the number of receptors expressed per cell and somatotropin release–inhibiting factor [SRIF]) may also
their responses are regulated, providing a further level of confer specific cellular response to a hormone and provide
control for hormone action. Several mechanisms account an additive cellular effect.10
for altered receptor function. Receptor endocytosis causes
internalization of cell surface receptors; the hormone-
receptor complex is subsequently dissociated, resulting in
abrogation of the hormone signal. Receptor trafficking may CONTROL OF HORMONE SECRETION
then result in recycling back to the cell surface (e.g.,
as for insulin), or the internalized receptor may undergo Anatomically distinct endocrine glands are composed of
lysosomal degradation. Both of these mechanisms, trig- highly differentiated cells that synthesize, store, and secrete
gered by activation of receptors, effectively lead to impaired hormones. Circulating hormone concentrations are a func-
hormone signaling by downregulation of these receptors. tion of glandular secretory patterns and hormone clearance
The hormone signaling pathway may also be downregu- rates. Hormone secretion is tightly regulated to attain cir-
lated by receptor desensitization (e.g., as for epinephrine); culating levels that are most conducive to eliciting the
ligand-mediated receptor phosphorylation leads to a revers- appropriate target tissue response. For example, longitudi-
ible deactivation of the receptor. Desensitization mecha- nal bone growth is initiated and maintained by exquisitely
nisms can be activated by a receptor’s ligand (homologous regulated levels of circulating GH: mild GH hypersecretion
desensitization) or by another signal (heterologous desen- results in gigantism, and GH deficiency causes growth
sitization), which attenuates receptor signaling in the con- retardation. Ambient circulating hormone concentrations
tinued presence of ligand. Receptor function may also be are not uniform, and the secretion patterns determine
limited by the action of specific phosphatases (e.g., SHP) appropriate physiologic function. For example, insulin
or by intracellular negative regulation of the signaling secretion occurs in short pulses elicited by nutrient intake
cascade (e.g., suppressor of cytokine signaling [SOCS] pro- and other signals; gonadotropin secretion is episodic,
teins inhibiting JAK-STAT signaling). determined by a hypothalamic pulse generator; and pro-
Mutational changes in receptor structure can also deter- lactin secretion appears to be relatively continuous with
mine hormone action. Constitutive receptor activation secretory peaks elicited during suckling.

Diseases caused by mutations in G-protein-coupled receptors


Condition Receptor Inheritance ∆ Function
Retinitis pigmentosa Rhodopsin AD/AR Loss
Nephrogenic diabetes insipidus Vasopressin V2 X-linked Loss
Isolated glucocorticoid deficiency ACTH AR Loss
Color blindness Red/green opsins X-linked Loss
Familial precocious puberty LH AD (male) Gain
Familial hypercalcemia Ca2+ sensing AD Loss
Neonatal severe parathyroidism Ca2+ sensing AR Loss
Dominant form hypocalcemia Ca2+ sensing AD Gain
Congenital hyperthyroidism TSH AD Gain
Resistance to thyroid hormone TSH AR (comp het) Loss
Hyperfunctioning thyroid adenoma TSH Somatic Gain
Metaphyseal chondrodysplasia PTH-PTHrP Somatic Gain
Hirschsprung’s disease Endothelin-B Multigenic Loss
Coat color alteration (E locus, mice) MSH AD/AR Loss and gain
Dwarfism (little locus, mice) GHRH AR Loss

Figure 1-3 Diseases caused by mutations in G-protein–coupled receptors. All are human conditions with the exception of the final two entries, which refer
to the mouse. Loss of function refers to inactivating mutations of the receptor, and gain of function to activating mutations. ACTH, adrenocorticotropic
hormone; AD, autosomal dominant; AR, autosomal recessive; LH, luteinizing hormone; TSH, thyroid-stimulating hormone; PTH-PTHrP, parathyroid hormone
and parathyroid hormone–related peptide; MSH, melanocyte-stimulating hormone; GHRH, growth hormone–releasing hormone; FSH, follicle-stimulating
hormone. (Reproduced from Mayo K. Receptors: molecular mediators of hormone action. In Conn PM, Melmed S, eds. Endocrinology: Basic and Clinical Principles.
Totowa, NJ: Humana Press, 1997:27.)
Principles of Endocrinology   9

Hormone secretion also adheres to rhythmic patterns. associated with declining slow-wave sleep and concomi-
Circadian rhythms serve as adaptive responses to environ- tant decline in GH and elevation of cortisol secretion.13
mental signals and are controlled by a circadian timing Most pituitary hormones are secreted in a circadian (day-
mechanism.11 Light is the major environmental cue adjust- night) rhythm, best exemplified by the ACTH peaks that
ing the endogenous clock. The retinohypothalamic tract occur before 9 a.m., whereas ovarian steroids follow a
entrains circadian pulse generators situated within hypo- 28-day menstrual rhythm. Disrupted episodic rhythms are
thalamic suprachiasmatic nuclei. These signals subserve often a hallmark of endocrine dysfunction. For example,
timing mechanisms for the sleep-wake cycle and determine loss of circadian ACTH secretion with high midnight cor-
patterns of hormone secretion and action. Disturbed circa- tisol levels is a feature of Cushing’s disease.
dian timing results in hormonal dysfunction and may also Hormone secretion is induced by multiple specific bio-
be reflective of entrainment or pulse generator lesions. For chemical and neural signals. Integration of these stimuli
example, adult GH deficiency due to a damaged hypothala- results in net temporal and quantitative secretion of the
mus or pituitary is associated with elevations in integrated hormone (Fig. 1-4). For example, signals elicited by hypo-
24-hour leptin concentrations, decreased leptin pulsatility, thalamic hormones (growth hormone–releasing hormone
and yet preserved circadian rhythm of leptin. GH replace- [GHRH], SRIF), peripheral hormones (IGF1, sex steroids,
ment restores leptin pulsatility, followed by loss of body thyroid hormone), nutrients, adrenergic pathways, stress,
fat mass.12 Sleep is also an important cue regulating and other neuropeptides all converge on the somatotroph
hormone pulsatility. About 70% of overall GH secretion cell, resulting in the ultimate pattern and quantity of GH
occurs during slow-wave sleep, and increasing age is secretion. Networks of reciprocal interactions allow for

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 1-4  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 trans-
ported 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 microgram amounts of specific anterior pituitary hormones. These enter the venous circulation through fenestrated local
capillaries, bind to specific target gland receptors, trigger release of micrograms 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 black dot at their origin indicate a secretory process. (Reproduced from Normal
AW, Litwack G. Hormones, ed 2. New York, NY: Academic Press, 1997:14.)
10   Principles of Endocrinology

dynamic adaptation and shifts in environmental signals. are secreted in nanogram amounts and have short half-
These regulatory systems embrace the hypothalamic pitu- lives of a few minutes. Anterior pituitary hormones are
itary and target endocrine glands as well as the adipocyte produced in microgram amounts and have longer half-
and lymphocyte. Peripheral inflammation and stress elicit lives, whereas peripheral hormones can be produced
cytokine signals that interface with the neuroendocrine in up to milligram amounts daily, with much longer
system, resulting in activation of the hypothalamic- half-lives.
pituitary axis. The parathyroid and pancreatic secreting A further level of secretion control occurs within the
cells are less tightly controlled by the hypothalamus, but gland itself. Intraglandular paracrine or autocrine growth
their functions are tightly regulated by the effects they peptides serve to autoregulate pituitary hormone secretion,
elicit. For example, PTH secretion is induced when serum as exemplified by EGF control of prolactin or IGF1 control
calcium levels fall, and the signal for sustained PTH of GH secretion. Molecules within the endocrine cell may
secretion is abrogated by rising calcium levels. also subserve an intracellular feedback loop. For example,
Several tiers of control play a part in the ultimate net corticotrope SOCS3 induction by gp130-linked cytokines
glandular secretion. First, central nervous system signals, serves to abrogate the ligand-induced JAK-STAT cascade,
including stress, afferent stimuli, and neuropeptides, blocking transcription of pro-opiomelanocortin and subse-
signal the synthesis and secretion of hypothalamic hor- quent secretion of ACTH. This rapid on-off regulation of
mones and neuropeptides (Fig. 1-5). Four hypothalamic- ACTH secretion provides a plastic endocrine response to
releasing hormones—GHRH, corticotropin-releasing changes in environmental signaling and serves to maintain
hormone, thyrotropin-releasing hormone, and homeostatic integrity.14
gonadotropin-releasing hormone (GnRH)—traverse the In addition to the central-neuroendocrine interface
hypothalamic portal vessels and impinge upon their mediated by hypothalamic chemical signal transduction,
respective transmembrane trophic hormone-secreting cell the central nervous system directly controls several hor-
receptors. These distinct cells express GH, ACTH, TSH, and monal secretory processes. Posterior pituitary hormone
gonadotropins, respectively. In contrast, hypothalamic secretion occurs as direct efferent neural extensions. Post-
somatostatin and dopamine suppress secretion of GH, pro- ganglionic sympathetic nerves also regulate rapid changes
lactin, and TSH. in renin, insulin, and glucagon secretion, and pregangli-
Trophic hormones also maintain the structural and onic sympathetic nerves signal to adrenal medullary cells,
functional integrity of endocrine organs, including the eliciting adrenaline release.
thyroid and adrenal glands and the gonads. Target hor-
mones, in turn, serve as powerful negative feedback regula-
tors of their respective trophic hormones; they often also
suppress secretion of hypothalamic-releasing hormones. In HORMONE MEASUREMENT
certain circumstances (e.g., during puberty), peripheral sex
steroids may positively induce the hypothalamic-pituitary– Endocrine function can be assessed by measuring levels of
target gland axis. LH induces ovarian estrogen secretion, basal circulating hormone, evoked or suppressed hormone,
which feeds back positively to induce further LH release. or hormone-binding proteins. Alternatively, peripheral
Pituitary hormones themselves, in a short feedback loop, hormone receptor function can be assessed. Meaningful
may also regulate their own respective hypothalamic- strategies for timing hormonal measurements vary from
controlling hormone. Hypothalamic releasing hormones system to system. In some cases, circulating hormone con-
centrations can be measured in randomly collected serum
samples. This measurement, when standardized for fasting,
environmental stress, age, and gender, is reflective of true
hormone concentrations only when levels do not fluctuate
CNS Inputs
appreciably. For example, thyroid hormone, prolactin, and
IGF1 levels can be accurately assessed in fasting morning
serum samples. If hormone secretion is clearly episodic,
Hypothalamus
Tier I timed samples may be required over a defined time course
Hypothalamic to reflect hormone bioavailability. For example, early
hormones morning and late evening cortisol measurements are most
appropriate. Sampling over 24 hours for GH measure-
ments, with samples collected every 2, 10, or 20 minutes,
is expensive and cumbersome, yet may yield valuable diag-
Tier II nostic information. Random sampling may reflect secre-
Pituitary Paracrine cytokines tion peaks or nadirs, confounding adequate interpretation
and growth factors
of results.
In general, confirmation of failed glandular function is
Pituitary trophic made by attempting to evoke hormone secretion by recog-
hormone
nized stimuli. For example, testing of pituitary hormone
Tier III
Target gland Peripheral
reserve may be accomplished by injecting appropriate
hormones hypothalamic releasing hormones. Injection of trophic
hormones, including TSH and ACTH, evokes specific target
Figure 1-5  Model for regulation of anterior pituitary hormone secretion gland hormone secretion. Pharmacologic stimuli (e.g.,
by three tiers of control. Hypothalamic hormones impinge directly on their metoclopramide for induction of prolactin secretion) may
respective target cells. Intrapituitary cytokines and growth factors regulate
tropic cell function through paracrine (and autocrine) control. Peripheral
also be useful to test hormone reserve. In contrast, hormone
hormones exert negative feedback inhibition on the synthesis and secretion hypersecretion can be diagnosed by suppressing glandular
of their respective pituitary trophic hormones. (Reproduced from Ray D, function. For example, failure to appropriately suppress
Melmed S. Pituitary cytokine and growth factor expression and action. Endocrine GH levels after a standardized glucose load implies inap-
Rev. 1997;18:206-228.) propriate GH hypersecretion. The failure to suppress insulin
Principles of Endocrinology   11

secretion in response to hypoglycemia indicates inappro- uncommonly, a host of genetic abnormalities can lead to
priate hypersecretion of insulin and should prompt a decreased hormone production. These disorders can result
search for the cause (e.g., an insulin-secreting tumor). from abnormal development of hormone-producing cells
Radioimmunoassays use highly specific antibodies (e.g., hypogonadotropic hypogonadism caused by KAL
unique to a hormone, or hormone fragment, to quantify gene mutations), from abnormal synthesis of hormones
hormone levels. Enzyme-linked immunosorbent assays (e.g., deletion of the GH gene), or from abnormal regula-
(ELISAs) employ enzymes instead of radioactive hormone tion of hormone secretion (e.g., the hypoparathyroidism
markers, and enzyme activity reflects hormone concentra- associated with activating mutations of the parathyroid
tion. This sensitive technique has allowed ultrasensitive cell’s calcium-sensing receptor).
measurements of physiologic hormone concentrations.
Hormone-specific receptors may be employed in place of
the antibody in a radioreceptor assay.
Altered Tissue Responses
Resistance to hormones can be caused by a variety of
genetic disorders. Examples include mutations in the GH
receptor in Laron dwarfism and mutations in the Gsα gene
ENDOCRINE DISEASES in the hypoparathyroidism of pseudohypoparathyroidism
type 1A. The insulin resistance in muscle and liver that is
Endocrine diseases fall into five broad categories: (1) central to the etiology of type 2 diabetes mellitus appears
hormone overproduction, (2) hormone underproduction, to be polygenic in origin. Type 2 diabetes is also an example
(3) altered tissue responses to hormones, (4) tumors of of a disease in which end-organ insensitivity is worsened
endocrine glands, and a relatively new category exempli- by signals from other organs, in this case by signals
fied in the thyroid axis, (5) hormone deficiency due to its originating in fat cells. In other cases, the target organ of
excessive rate of inactivation caused by overexpression of hormone action is more directly abnormal, as in the PTH
an endogenous enzyme in a tumor. resistance of renal failure.
Increased end-organ function can be caused by muta-
tions in signal reception and propagation. For example,
Hormone Overproduction activating mutations in TSH, LH, and PTH receptors can
Occasionally, hormones are secreted in increased amounts cause increased activity of thyroid cells, Leydig cells, and
because of genetic abnormalities that cause abnormal regu- osteoblasts, even in the absence of ligand. Similarly, acti-
lation of hormone synthesis or release. For example, in vating mutations in the Gsα protein can cause precocious
glucocorticoid-remediable hyperaldosteronism, an abnor- puberty, hyperthyroidism, and acromegaly in McCune-
mal chromosomal crossover event puts the aldosterone Albright syndrome.
synthetase gene under the control of the ACTH-regulated
11β-hydroxylase gene. More often, diseases of hormone
overproduction are associated with an increase in the total
Tumors of Endocrine Glands
number of hormone-producing cells. For example, the Tumors of endocrine glands, as mentioned earlier, often
hyperthyroidism of Graves’ disease, in which antibodies result in hormone overproduction. Some tumors of endo-
mimic TSH and activate the TSH receptors on thyroid cells, crine glands produce little if any hormone but cause disease
is associated with a dramatic increase in thyroid cell pro- through local compressive symptoms or metastatic spread.
liferation, as well as increased synthesis and release of Examples include so-called nonfunctioning pituitary
thyroid hormone from each thyroid cell. The increase in tumors, which are usually benign but can cause a variety
thyroid cell number represents a polyclonal expansion of symptoms due to compression on adjacent structures,
of thyroid cells in which large numbers of thyroid cells and thyroid cancer, which can spread throughout the body
proliferate in response to an abnormal stimulus. However, without causing hyperthyroidism.
most endocrine tumors are not polyclonal expansions but
instead represent monoclonal expansions of one mutated
cell. Pituitary and parathyroid tumors, for example, are Excessive Hormone Inactivation
usually monoclonal expansions in which somatic muta-
tions in multiple tumor suppressor genes and proto-
or Destruction
oncogenes occur. These mutations lead to increased Although most enzymes important for endocrine systems
proliferation or survival (or both) of the mutant cells. activate a prohormone or precursor protein, there are also
Sometimes, this proliferation is associated with abnormal those whose function is to inactivate the hormone in a
secretion of hormone from each tumor cell. For example, physiologically regulated fashion. An example is iodothy-
mutant Gs α-subunit proteins in somatotrophs can lead to ronine deiodinase type 3 (D3) which inactivates T3 and T4
both increased cellular proliferation and increased secre- by removing an inner-ring iodine atom from the
tion of GH from each tumor cell. iodothyronine. The products of these reactions, 3,3′-
diiodothyronine and reverse T3, respectively, are inactive.
Several years ago, an infant was identified with hypothy-
Hormone Underproduction roidism due to a large hepatic hemangioma expressing
Underproduction of hormone can result from a wide high amounts of D3. This condition was termed consump-
variety of processes, ranging from surgical removal of para- tive hypothyroidism because it resulted from inactivation of
thyroid glands during neck surgery, to tuberculous destruc- thyroid hormone at a more rapid rate than it could be
tion of adrenal glands, to iron deposition in beta cells of produced by the infant’s normal thyroid gland.15,16 The
islets in hemochromatosis. A frequent cause of destruction condition has now been identified in a number of infants
of hormone-producing cells is autoimmunity. Autoim- and even in adults with D3-expressing tumors. In theory,
mune destruction of beta cells in type 1 diabetes mellitus accelerated destruction of other hormones could occur
or of thyroid cells in Hashimoto’s thyroiditis are two of the from similar processes, but there are no examples reported
most common disorders treated by endocrinologists. More to date.
12   Principles of Endocrinology

require frequent administration by injection and close


THERAPEUTIC STRATEGIES monitoring by the patient. Novel treatment systems that
would link frequent monitoring of glucose levels to
In general, hormones are employed pharmacologically for appropriate adjustments in the insulin dose promise to
their replacement or suppressive effects. Hormones may substantially reduce the burden of this disease. Hormones
also be used for diagnostic stimulatory purposes (e.g., are biologically powerful molecules that exert therapeutic
hypothalamic hormones) to evoke target organ responses benefit and effectively replace pathologic deficits. They
or to diagnose endocrine hyperfunction by suppressing should not be prescribed without clearcut indications and
hormone hypersecretion (e.g., T3). Ablation of endocrine should not be administered without careful evaluation by
gland function from genetic or acquired causes can be an appropriately qualified medical practitioner.
restored by hormone replacement therapy. In general,
steroid and thyroid hormones are replaced orally, whereas
peptide hormones (e.g., insulin, GH) require injection.
Gastrointestinal absorption and first-pass kinetics deter- REFERENCES
mine oral hormone dosage and availability. Physiologic 1. Liu PT, Stenger S, Li H, et al. Toll-like receptor triggering of
replacement can achieve both appropriate hormone levels a vitamin D-mediated human antimicrobial response. Science.
(e.g., thyroid) and approximate hormone secretory 2006;311:1770-1773.
2. Chawla A, Repa JJ, Evans RM, et al. Nuclear receptors and lipid physiol-
patterns (e.g., GnRH delivered intermittently via a pump). ogy: opening the X-files. Science. 2001;294:1866-1870.
Hormones can also be used to treat diseases associated 3. Mendel CM, Weisiger RA, Jones AL, et al. Thyroid hormone-binding
with glandular hyperfunction. Long-acting depot prepara- proteins in plasma facilitate uniform distribution of thyroxine
tions of somatostatin analogues suppress GH hypersecre- within tissues: a perfused rat liver study. Endocrinology. 1987;120:
1742-1749.
tion in acromegaly or 5-hydroxyindoleacetic acid (5-HIAA) 4. Mendel CM. The free hormone hypothesis: physiologically based math-
hypersecretion in carcinoid syndrome. Estrogen receptor ematical model. Endocr Rev. 1989;10:232-274.
antagonists (e.g., tamoxifen) are useful for some patients 5. Safadi FF, Thornton P, Magiera H, et al. Osteopathy and resistance to
with breast cancer, and GnRH analogues may downregu- vitamin D toxicity in mice null for vitamin D binding protein. J Clin
Invest. 1999;103:239-251.
late the gonadotropin axis and benefit patients with pros- 6. Palha JA, Fernandes R, de Escobar GM, et al. Transthyretin regulates
tate cancer. thyroid hormone levels in the choroid plexus, but not in the brain
Novel formulations of receptor-specific hormone ligands parenchyma: study in a transthyretin-null mouse model. Endocrinology.
are now being clinically developed for more selective 2000;141:3267-3272.
7. Palha JA, Episkopou V, Maeda S, et al. Thyroid hormone metabolism
therapeutic targeting; examples include estrogen agonists/ in a transthyretin-null mouse strain. J Biol Chem. 1994;269:33135-
antagonists and somatostatin receptor subtype ligands. 33139.
Modes of hormone injection (e.g., for PTH) may also 8. Visser WE, Friesema EC, Jansen J, et al. Thyroid hormone transport by
determine therapeutic specificity and efficacy. Improved monocarboxylate transporters. Best Pract Res Clin Endocrinol Metab.
2007;21:223-236.
hormone delivery systems, including computerized mini- 9. Hammes A, Andreassen TK, Spoelgen R, et al. Role of endocytosis in
pumps, intranasal sprays (e.g., for desmopressin [DDAVP]), cellular uptake of sex steroids. Cell. 2005;122:751-762.
pulmonary inhalations, and depot intramuscular injec- 10. Rocheville M, Lange DC, Kumar U, et al. Receptors for dopamine and
tions, allow added patient compliance and ease of admin- somatostatin: formation of hetero-oligomers with enhanced functional
activity. Science. 2000;288:154-157.
istration. Insulin delivered by inhalation has already been 11. Moore RY. Circadian rhythms: basic neurobiology and clinical applica-
approved for use, and inhaled forms of GH and other tions. Ann Rev Med. 1997;48:253-266.
hormones are under investigation. Cell-based therapies 12. Aftab MA, Guzder R, Wallace AM, et al. Circadian and ultradian rhythm
employing highly programmed pluripotent stem cells and leptin pulsatility in adult GH deficiency: effects of GH replacement.
J Clin Endocrinol Metab. 2001;86:3499-3506.
expressing a particular hormone or growth factor are also 13. Cauter EV, Leproult R, Plat L. Age-related changes in slow wave sleep
in development These approaches require novel adminis- and REM sleep and relationship with growth hormone and cortisol
tration systems to allow cell-derived endocrine products to levels in healthy men. JAMA. 2000;284:861-868.
reach their intended target.17 Novel technologies poten- 14. Melmed S. The immuno-neuroendocrine interface. J Clin Invest.
2001;108:1563-1566.
tially marked prolongation in the half-life of peptide hor- 15. Huang SA, Tu HM, Harney JW, et al. Severe hypothyroidism caused by
mones, which would require less frequent administration. type 3 iodothyronine deiodinase (D3) in infantile hemangiomas.
For example, a once-weekly preparation of exenatide, a N Engl J Med. 2000;343:185-189.
GLP1 analogue used in the treatment of type 2 diabetes as 16. Bianco AC, Salvatore D, Gereben B, et al. Biochemistry, cellular and
molecular biology, and physiological roles of the iodothyronine
a twice-daily injection, is undergoing clinical trials. selenodeiodinases. Endocrine Rev. 2002;23:38-89.
Despite this tremendous progress, some therapies, such 17. Graf T, Enver T. Forcing cells to change lineages. Nature. 2009;
as insulin delivery to rigorously control blood sugar, still 462:587-594.

You might also like