Sie sind auf Seite 1von 17

n n

2n n n n n
ENDOCRINE FUNCTION OF THE
GASTROINTESTINAL TRACT
n n n n n n n n

O B J E C T I V E S
1. Understand the role of well-established GI hormones n The regulation of secretion from the exocrine pancreas
associated with the following four major aspects of and the gallbladder and their associated ducts
GI physiology: n The stimulation of GI tract growth (an enterotropic
n The regulation of gastric acid secretion and gastric action)
motility n The enhancement of nutrient-induced insulin secre-
tion by the endocrine pancreas (incretin action)

Note: A fifth general function of GI hormones, the effect on appetite, is discussed in the context of energy homeostasis in Chapter 3.

W e begin our discussion of endocrine


physiology with the hormonal function and regulation
of the gastrointestinal (GI) tract. The discovery of se-
or transporters that allow the cell to sample the
contents of the lumen. Luminal contents, called secre-
togogues, stimulate specific enteroendocrine cell types
cretin in 1902 by Bayliss and Starling represented the to secrete their hormones. This sampling or nutrient
first characterization of a hormone as a blood-borne tasting is independent of osmotic and mechanical
chemical messenger, released at one site and acting forces. The secretogogue mechanisms involved are
at multiple other sites. Indeed, the epithelial layer of poorly understood, but some appear to require the ab-
the mucosa of the GI tract harbors numerous enter- sorption of the nutrient. There is also evidence for the
oendocrine cell types, which collectively represent luminal secretion of paracrine peptide factors from
the largest endocrine cell mass in the body. the surrounding absorptive epithelial cells that stimu-
The diffuse enteroendocrine system is perhaps the late hormonal release from enteroendocrine cells. As
most basic example of endocrine tissue in that it is part of their response to luminal contents, specific
composed of unicellular glands situated within a sim- enteroendocrine cell types display distinct localiza-
ple epithelium. Most enteroendocrine cells, called tions along the GI tract (Table 2-1). We will see that
open cells, extend from the basal lamina of this epithe- these localizations are central to the regulation and
lium to the apical surface (Fig. 2-1), although there are function of each cell type.
also closed enteroendocrine cells, which do not ex- In the simplest model of enteroendocrine cell func-
tend to the luminal surface. The apical membranes tion, a hormone is released from the basolateral mem-
of open enteroendocrine cells express either receptors brane in response to the presence of a secretogogue at
27
28 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

1. Specific luminal secretogogues


(e.g., H+, amino acids, fatty acids)
stimulate “open” enteroendocrine cell at apical surface

Epithelium of mucosa
that lines the lumen
of the GI tract
FIGURE 2-1 n Closed and open
“Closed” type cell
enteroendocrine cells. Entero- Also regulated by Open Closed • Does not reach lumen
endocrine cells sit within the neuronal, paracrine, • Must be regulated by
simple epithelium of the GI and endocrine inputs neuronal, paracrine, or
tract. “Open” cells extend from endocrine inputs
the basal lamina to the lumen. 2. “Open” and “closed”
cells secrete hormone 3. Hormone enters
“Closed” cells do not reach splanchnic blood vessels
the lumen. Both cells secrete from basolateral side
in lamina propria
hormones that enter capillaries
in the lamina propria beneath
the epithelium.

Arterial supply Venous return


(to the hepatic portal vein)

the luminal side of the cell. The secreted hormone dif- luminal concentration of its secretogogue diminishes,
fuses into blood vessels in the underlying lamina pro- thereby terminating the secretion of the hormone.
pria, thereby gaining access to the general circulation. This simple model of the enteroendocrine system
Circulating GI hormones regulate GI tract functions does not fully account for the integration with other
by binding to specific receptors at one or more sites systemic responses to a meal. Both open and closed
within the GI tract and its extramural glands. In the enteroendocrine cells are regulated by the enteric ner-
classic model, the secretion of the hormone by an enter- vous system (ENS) and paracrine factors secreted by
oendocrine cell is subsequently terminated when the neighboring epithelial cells (intrinsic regulators of

TABLE 2-1
Distribution of Enteroendocrine Cells Along the GI Tract
STOMACH DUODENUM JEJUNUM ILEUM COLON

G cell (gastrin) x (x)


S cell (secretin) x x
I cell (CCK) x x (x)
K cell (GIP) x x
L cell (GLP-1) x x
L cell (GLP-2) x x
M cell (motilin) x x
Ghrelin-secreting cell x (x) (x) (x) (x)

x, Primary location of concentration; (x), less concentrated.


ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 29

enteroendocrine cell function). Additionally, there are


ENTEROENDOCRINE HORMONE
extrinsic regulators of enteroendocrine cells, most
FAMILIES AND THEIR RECEPTORS
notably the autonomic nervous system and endocrine All established GI hormones are peptides and bind to
glands that reside outside of the GI tract. Conversely, GI G-protein-coupled receptors (GPCRs; see Chapter 1)
hormones can have local (i.e., paracrine) actions on the located on the plasma membrane of target cells. GI
afferent nerves of autonomic or enteric reflexes, so hormones, as well as their cognate GPCRs, can be or-
the response to a GI hormone can be mediated by a ganized into gene families based on structural homol-
neurotransmitter. Thus, GI tract function is orches- ogies. In this chapter, we discuss members of three
trated through a complex interplay of neural and endo- enteroendocrine hormone families: gastrin, secre-
crine responses and actions. It is not surprising, tin, and motilin (Table 2-2).
therefore, that GI function is often perturbed in pa- The gastrin family includes gastrin and cholecys-
tients with psychiatric disorders (e.g., depression) and tokinin (CCK), which share a common stretch of 5
endocrine disorders (e.g., hyperthyroidism). amino acids at the C-terminus. Gastrin binds with
The hormones secreted by the enteroendocrine sys- high affinity to the CCK-2 receptor (previously called
tem function to maintain the health of the GI tract and the CCK-B/gastrin receptor). CCK binds with high
its extramural glands and provide an integrated re- affinity to the CCK-1 receptor.
sponse to the acquisition of nutrients. This integrated The secretin family includes the hormones secretin,
response to GI hormones is due, in part, to their ability glucagon, and glucagon-like peptides (including
to regulate multiple functions of the GI tract. GLP-1 and GLP-2) and gastric inhibitory polypeptide

TABLE 2-2
Enteroendocrine Hormone Families and Their Receptors
RECEPTOR AND PRIMARY PRIMARY DISTRIBUTION OF THE RECEPTOR
HORMONE FAMILY MEMBERS OF FAMILY SIGNALING PATHWAY (RELATED TO GI FUNCTION)

Gastrin Gastrin (G cell) CCK2 receptor Gastric ECL cell and parietal cell
Gq - " in Ca2 þ and PKC
CCK (I cell) CCK1 receptor Gallbladder muscularis and sphincter of
Gq - " in Ca2 þ and PKC hepatopancreatic ampulla
Pancreatic acinar cells
Pancreatic ducts
Vagal afferents and enteric neurons
Stomach muscularis and pyloric sphincter
Gastric D cells
Secretin Secretin (S cell) Secretin receptor Pancreatic ducts and biliary ducts
Gs - " in cAMP Pancreatic acinar cells
G cells and pancreatic cells
GLP-1 (L cell) GLP-1 receptor b Cells of pancreatic islets
Gs - " in cAMP
GLP-2 (L cell) GLP-2 receptor GI tract, especially small intestine
Gs - " in cAMP
GIP (K cell) GIP receptor b Cells of pancreatic islets
Gs - " in cAMP Gastric mucosa and muscularis
Motilin Motilin (M cell) Motilin receptor Stomach and small intestines, especially in smooth
Gq - " in Ca2 þ and PKC muscle cells and enteric neurons
(also binds erythromycin)
Ghrelin (P/D1 cell) GHS receptor type 1a (GHS-RIa) Pituitary and hypothalamus
Gq - " in Ca2 þ and PKC
30 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

(GIP; more recently referred to as glucose-dependent the small intestine, the duodenum. The layers of the
insulinotropic peptide—see later). This family also stomach wall carry out two basic functions: secretion
includes the neurocrine factor, vasoactive intestinal and contraction/relaxation.
peptide (VIP). The corresponding GPCRs for each
member of the secretin family of peptides are also struc-
turally related. These receptors are all primarily coupled Overview of Regulation of Gastric
to Gs signaling pathways that increase intracellular cy- Secretion and Motility
clic adenosine monophosphate (cAMP) in target cells. The innermost layer of the stomach wall, the gastric
The motilin family includes the hormones motilin mucosa, contains glandular and surface mucus-
and ghrelin. Ghrelin was originally identified as a producing epithelia and can be divided into proximal
growth hormone secretogogue (GHS) but is most and distal segments. Two of the proximal portions of
abundant in the fundus of the stomach. The receptors the stomach (fundus and body) contain the main gas-
for motilin and ghrelin are GPCRs that are linked to tric mucosal glands (Fig. 2-2). Within these glands, the
Ga-q/phospholipase/IP3 pathways, which, in turn, parietal cells secrete HCl, which is important for hy-
stimulate protein kinase C- and Ca2 þ-dependent sig- drolysis of macromolecules, activation of proenzymes,
naling pathways (see Chapter 1). and the sterilization of ingested food. Parietal cells
Many GI peptides are also expressed by tissues also secrete intrinsic factor, which is a glycoprotein
outside of the GI tract. Pathophysiologically, GI pep- required for the efficient absorption of vitamin B12.
tides can be secreted in an uncontrolled manner from The glands of the fundus and body also contain the
tumors. Other physiologic sites of production include chief cells, which secrete digestive enzymes (e.g., pep-
other endocrine glands (e.g., the pituitary gland) and sinogen, gastric lipase). A third cell type, the mucous
reproductive structures. Several peptides are produced cell, is found in the neck of the gastric glands and on
by the central (CNS) and peripheral (PNS) nervous the surface throughout the stomach. Mucous cells se-
systems, where they are used as neurotransmitters or crete mucigens, which buffer and protect the lining of
neuromodulatory factors. For example, cholecystoki- the stomach, particularly in the vicinity of the main
nin (CCK) is expressed in the neocortical region of the gastric glands. Because gastric enzyme and mucus pro-
CNS and the genitourinary-associated nerves of the duction is primarily under nervous control, with little
PNS. As for its role in the CNS, CCK has been linked endocrine input, we focus here on gastric acid secre-
to anxiety and panic disorders. This also means that tion and motility.
receptors for these peptides also reside within the The distal part of the gastric mucosa, the pyloric
CNS, the PNS, and probably other non-neural tissues. antrum, has an important enteroendocrine function.
Thus, a pharmacologic agent (agonist or antagonist) This part of the stomach contains two types of “open”
related to a specific GI peptide can potentially have enteroendocrine cells. The G cells secrete gastrin, a
a wide range of effects, depending on its stability hormone, and the D cells secrete somatostatin, a
and whether it can cross the blood-brain barrier. paracrine factor. These two peptides act antagonisti-
The possibility also exists that extra-GI sites of synthe- cally in a negative feedback loop to regulate gastric
sis can “spill over” into the general circulation and blood flow, cell growth, secretion, and motility (see
affect GI function. later). D cells are also found within the fundus and
body region, where they directly inhibit parietal cell
secretion.
GASTRIN AND THE REGULATION
An outer layer of the stomach wall, the muscularis
OF GASTRIC FUNCTION
externa, is composed of smooth muscle. The relaxa-
The stomach acts as a food reservoir. People eat dis- tion of this muscle allows distention and storage,
continuously and typically eat more at one sitting than and its contractions ultimately move the partially
their GI tract can process immediately. Thus, the digested food (chyme) into the duodenum. There
stomach holds the ingested food and gradually releases are two gateways into and out of the stomach. These
partially digested food (chyme) into the first part of are the lower esophageal sphincter (LES) and the
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 31

Fundus of stomach
Lower esophageal
sphincter

Main gastric glands


• Site of acid secretion and
intrinsic factor secretion
from parietal cells
• Site of enzyme secretion
from chief cells Body of
stomach
Pyloric sphincter FIGURE 2-2 n Anatomy of the stomach.
Antrum of stomach
• Site of gastrin
secretion from
G cells

Antrum of stomach

Duodenum

pyloric sphincter, respectively. The LES allows swal- although patients with a gastrin-producing tumor
lowed food particles to enter the stomach and protects (Zollinger-Ellison syndrome) can present with ulcer-
the esophagus from the reflux of acidic chyme. The ation of the esophagus, stomach, and duodenum.
pyloric sphincter operates in conjunction with the The general model of gastric control in response to a
muscularis externa to allow only small particles of meal can be organized into three phases. The cephalic
digested chyme to escape the stomach and enter phase, which accounts for about 20% of the response to
the duodenum. The pyloric sphincter also prevents a meal during the digestive period, is activated by the
backflow of chyme into the stomach. actual or imagined smell and sight of food, or by the
In general, regulation of gastric function involves presence of food in the mouth. The cephalic phase is
the stimulation of secretion and motility as needed associated with increased gastric secretion but de-
(i.e., in the presence of food), and the inhibition of creased motility, in anticipation of the need to store
gastric secretion and motility as acidic chyme reduces and start digesting food. The gastric phase, which ac-
the pH of the stomach, or as chyme moves into the counts for about 10% of the postprandial response, is
small intestine and colon. In this way, the stomach activated by the presence of food in, and mechanical
avoids excessive acid secretion in the absence of buff- distention of, the stomach. During the gastric phase, se-
ering foodstuffs. Further, the portion of the GI tract cretion is strongly stimulated, and this is accompanied
below the stomach protects itself from exposure to ex- by an increase in peristaltic contractions and gastric
cessive amounts of acid, which is both damaging to the emptying. The third phase is the intestinal phase, dur-
intestinal lining and inhibitory to the activity of intes- ing which an acidic mixture of partially digested food
tinal enzymes. Additionally, the small intestine, in (chyme) moves in a regulated manner through the py-
which the majority of digestion and absorption oc- loric sphincter into the small intestine and ultimately
curs, controls the flow rate of food into and through into the colon. The processes of enzymatic digestion
the small intestines in order to optimize digestion and absorption that occur during the digestive phase
and absorption of nutrients, salts, and water. The in- account for 70% of the digestive period. The movement
ability to properly regulate acid secretion and its flow of food into the lower GI tract generally moderates both
into the intestine usually gives rise to duodenal ulcers, gastric secretion and emptying.
32 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

Gastrin and the Stimulation of granules that generates C-termini peptides. Processing
Gastric Function of these intermediates also includes the cyclization of
Gastric HCl secretion from parietal cells is stimulated the glutaminyl to a pyroglutamyl residue. The third
by three pathways: stage involves the amidation of the C-terminus to pro-
duce amidated gastrins. The primary secreted bioac-
n Paracrine stimulation by histamine, which is tive product of human G cells is G-17 (i.e., 17
secreted by neighboring enterochromaffin-like amino acids). The pyroglutamyl residue at the amino
(ECL) cells terminus and the amidation of the C-terminus protect
n Enteric nervous system and vagal parasympa- G-17 from digestion by circulating aminopeptidases
thetic nervous system stimulation via gastrin- and carboxypeptidases. G-17 binds with high affinity
releasing peptide (GRP) and acetylcholine to the CCK2 receptor and is responsible for all of the
n Direct and indirect hormonal stimulation by the gastrin effects on the stomach. The last four amino
peptide hormone gastrin acids assign gastrin-like biologic activity to G-17. A
Gastrin is produced by the G cells of the stomach synthetic, clinically used form of gastrin, pentagas-
antrum and proximal duodenum. In humans, the trin, contains the last four amino acids, plus an ala-
term gastrin refers to a 17-amino acid peptide that nine at the amino terminus that confers increased
has modifications at both termini (G-17). In fact, stability.
the production of G-17 is an excellent example of During the cephalic phase, gastric HCl secretion is
how a peptide-encoding gene gives rise to multiple, stimulated by vagal (parasympathetic) inputs. Pre-
larger precursors, which are also secreted into the ganglionic vagal efferents activate enteric neurons
blood. G-17 is the product of sequential posttransla- that directly stimulate the parietal cells and stimulate
tional processing of preprogastrin, which can be gen- the release of histamine from ECL cells (Fig. 2-4).
erally characterized in three phases (Fig. 2-3). In the These actions are mediated by acetylcholine, which
first phase, sulfation and proteolysis generate a mix- binds to the muscarinic receptor. Vagal stimulation
ture of gastrin precursors, called progastrins. The of gastrin is mediated by the neurocrine factor, GRP,
second phase involves proteolysis within secretory released from enteric neurons.

Preprogastrin

• Sulfation of tyrosyl residue near C-terminus


• Endoproteolysis into several fragments

Progastrins

• Further endoproteolysis
FIGURE 2-3 n Processing of • Cyclization of glutaminyl residue at N-terminus to a pyroglutamyl
preprogastrin. residue

Gastrin intermediates

• Further endoproteolysis of larger fragments into 17-amino acid


C-terminal peptide, which is primary product of G cells in humans
• Amidation of C-terminus

Bioactive 17-amino
acid gastrin (G-17)
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 33

To heart Gastrin in hepatic portal vein FIGURE 2-4 n Regulation of


gastric HCl secretion during the
Preganglionic
Enteric cephalic phase of a meal. The
vagal
ACh E GRP G cell thought, sight, or smell of food,
or the presence of food in the
mouth, stimulates acid secretion
through the vagal preganglionic
Preganglionic parasympathetic nerves, which
vagal Enteric Parietal stimulate the release of acetylcho-
ACh E ACh H⫹ line (ACh) from postganglionic
cell
enteric nerves. Enteric nerve fibers
secreting ACh stimulate parietal
HIST
cells directly and through the
release of histamine from entero-
ECL chromaffin-like (ECL) cells. Gas-
cell trin is also stimulated by enteric
Preganglionic
neuronal fibers that release gastrin-
vagal Enteric releasing peptide (GRP). As a hor-
ACh E ACh mone, gastrin levels increase in the
GAST general circulation. Gastrin stimu-
lates gastric HCl secretion by bind-
From heart Gastrin in gastric veins ing to CCK2 receptors on ECL
cells (and, to a lesser extent, on
Lamina propria Epithelium Gastric
lumen parietal cells).

During the gastric phase, gastrin secretion from G leads to exocytosis of histamine-containing secretory
cells is primarily stimulated by the presence of pep- vesicles. Gastrin also increases histamine synthesis
tides and amino acids in the lumen of the antrum and storage by increasing the expression of histidine
(Fig. 2-5). Gastrin secretion can also be stimulated decarboxylase, which generates histamine from histi-
by stomach distention as detected by mechanosen- dine, and type 2 vesicular monoamine transporter
sors during the gastric phase, acting through local (VMAT-2), which transports and concentrates hista-
neuronal pathways, and through a vagovagal reflex. mine into the secretory vesicles. Thus, gastrin coordi-
Circulating gastrin levels increase by several-fold nates both the secretion and synthesis of histamine in
within 30 to 60 minutes after ingestion of a meal. ECL cells. Histamine, in turn, stimulates HCl secretion
The primary action of gastrin is the stimulation of in a paracrine manner by binding to the H2 receptor
HCl secretion by the parietal cells of the gastric glands on nearby epithelial parietal cells. Gastrin also has a
within the fundus and body of the stomach. To accom- direct, although less important, effect on parietal cells.
plish this, gastrin must enter and circulate through the During the intestinal phase of a meal, the decrease
general circulation and then exit capillaries and ven- in gastric contents relieves the stimulation of G cells by
ules within the lamina propria of the gastric mucosa amino acids and peptides, and by distention-induced
in the body and fundus (i.e., upstream of where gastrin vagovagal pathways. The decrease in gastric contents
is released within the stomach). also reduces the buffering capacity of the gastric lu-
Gastrin evokes HCl secretion primarily through men. Thus, during the intestinal phase and the inter-
binding to the CCK2 receptor on ECL cells. ECL cells, digestive period, the acidity of the stomach decreases.
which reside in the lamina propria of the gastric mu- When the pH falls below 3, acid stimulates the D cells
cosa, produce histamine in response to gastrin (see to secrete the paracrine peptide, somatostatin. So-
Fig. 2-5). Gastrin binding to the Gq-coupled CCK2 re- matostatin acts through its receptors (SS-R) to inhibit
ceptor on ECL cells increases intracellular Ca2 þ, which gastrin secretion from neighboring G cells (Fig. 2-6).
34 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

To heart Gastrin in hepatic portal vein

Preganglionic
vagal Enteric
ACh E GRP AAs
G cell Peptides

Chyme
FIGURE 2-5 n Regulation of Preganglionic
gastrin secretion during the vagal Enteric Parietal
gastric phase of a meal. Luminal ACh E ACh H⫹
cell
amino acids and peptides
strongly stimulate G cells in
Afferent of HIST
the antrum to secrete gastrin.
vagovagal response
Gastrin secretion and HCl secre- Mechanosensors
tion are also stimulated by stom- ECL
ach distention through local and cell
autonomic (vagovagal) reflexes. Preganglionic
vagal Enteric
ACh E ACh
GAST

From heart Gastrin in gastric veins

Lamina propria Epithelium Gastric


lumen

G cell
FIGURE 2-6 n Regulation of gas-
trin secretion during the intestinal
phase of a meal. The exit of food
(chyme) from the stomach lumen SS
reduces buffering of HCl. A low
pH stimulates D cells to release
the paracrine factor, somato-
statin (SS), which inhibits gastrin
Enterogastrones from
secretion from neighboring G
intestines in gastric D cell H⫹
cells. The exact nature of physio- veins
logic enterogastrones in humans
is not well established. Candi-
dates include secretin and gastric- Parietal
inhibitory peptide (gip) from the cell
small intestine, and peptide yy ECL
cell
from the ileum and colon.

Lamina propria Epithelium Gastric


lumen
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 35

Gastrin release and gastric emptying are also


inhibited during the intestinal phase by the release
ENTEROENDOCRINE REGULATION
of hormones and neural signals from the small
OF THE EXOCRINE PANCREAS
intestine and colon in response to acidity, hyperto-
AND GALLBLADDER
nicity, distention, and specific molecules (e.g., fatty The exocrine pancreas is an extramural gland that
acids). These hormones are collectively referred to empties its secretory products through a main ex-
as enterogastrones. The identity of the physiologic cretory duct into the GI tract at the duodenum
enterogastrones in humans that inhibit gastric acid (Fig. 2-7). The acini of the exocrine pancreas produce
secretion remains uncertain but includes candidates enzymes necessary to digest macromolecules in the
such as secretin and GIP from the duodenum small intestine. Pancreatic enzymes have optimal ac-
and jejunum and peptide YY and GLP-1 from the tivities at a neutral pH. Accordingly, the cells that line
distal ileum and colon. CCK is a well-established the pancreatic ducts secrete a bicarbonate-rich fluid,
inhibitor of gastric motility and emptying. CCK which serves to neutralize acidic chyme in the duode-
is released from the duodenum and jejunum in num. The gallbladder is also an extramural organ. It
response to the presence of luminal fatty acids (see receives bile that is secreted by the liver. Bile is both
Fig. 2-6). stored and concentrated in the gallbladder. Bile is

Bile duct

Accessory pancreatic duct

Minor duodenal papilla Main pancreatic duct

Major duodenal papilla

Hepatopancreatic ampulla

FIGURE 2-7 n Anatomy of the common bile duct, pancreas, pancreatic duct, and duodenum. The gallbladder (not shown)
stores and concentrates bile from the liver. Contraction of the gallbladder and relaxation of the sphincter of Oddi
(surrounds the hepatopancreatic ampulla) allows bile to flow down the common bile duct into the duodenum.
Pancreatic enzymes and bicarbonate reach the duodenum via larger and larger ducts that eventually form the main
pancreatic duct. This duct joins the common bile duct just before it reaches the duodenum, to form the
hepatopancreatic ampulla. Inset shows a higher magnification of the exocrine pancreas. The termini of the secretory
units are the pancreatic acini, which secrete enzymes. The ductal epithelium secretes a bicarbonate-rich fluid. Note
that the ductal epithelium of the common bile duct also secretes a bicarbonate-rich fluid. (ª Elsevier. Drake et al:
Gray’s Anatomy for Students, www.studentconsult.com.)
36 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

released into small intestine through the common bile acinar cells and gallbladder contraction. This dual reg-
duct, which usually joins the main pancreatic duct to ulation allows for fine-tuning of the qualitative nature
form the hepatopancreatic ampulla just before open- of the product (e.g., in terms of the percentage of
ing into the duodenum (see Fig. 2-7). A major func- bicarbonate and protein in pancreatic juice) that is
tion of bile is the emulsification of triglycerides finally secreted into the duodenum.
to increase their accessibility to pancreatic lipase. The classic model for secretin and CCK action on
In order to perform this function, aggregates (called the pancreas is that the appearance of acid, long-chain
micelles) of bile acids and other lipids are required. fatty acids, and glycine-containing dipeptides and
Micelle formation requires neutral or slightly tripeptides in the duodenum stimulates the open
alkaline conditions. Accordingly, the epithelial cells enteroendocrine cells to secrete the two hormones.
of the common bile duct secrete a bicarbonate-rich Secretin and CCK then circulate in the blood and bind
fluid. to their specific receptors on either ductal or acinar
Pancreatic and gallbladder functions are primarily cells, respectively (Fig. 2-8).
regulated by the autonomic nervous system during However, there is evidence that secretin has
the interdigestive period (pancreatic secretion occurs permissive effects on CCK actions, and vice versa.
in phase with the migrating myoelectric complex Moreover, it is also clear that the autonomic and en-
[MMC] in humans), and during the cephalic and gas- teric nervous systems have a permissive effect on the
tric phases of the digestive period. However, during secretin and CCK actions. The neurotransmitter,
the intestinal phase, when these glands are most ac- ACh, and a secretin-related enteric neurocrine pep-
tive, they are predominantly under endocrine control tide, VIP, stimulate pancreatic ductal and acinar cells
by two GI hormones, secretin and CCK. Secretin pri- and synergize with secretin and CCK. Patients who
marily regulates ductal secretion of a bicarbonate-rich have a VIPoma (i.e., a tumor producing high levels
fluid from both pancreatic and bile ducts. CCK pri- of VIP) suffer from pancreatic diarrhea because of a
marily stimulates enzyme secretion from pancreatic constant high level of pancreatic secretion into the gut.

Duodenal
lumen
Pancreatic juice
reaches the duodenum
Secretin S cell H+ via the pancreatic duct
and hepatopancreatic
ampulla

Fatty acids,
CCK I cell dipeptides,
FIGURE 2-8 n Hormonal regula- tripeptides
tion of pancreatic secretion by
secretin and CCK.

CCK1R

Pancreatic enzymes
Bicarbonate-rich fluid

Secretin-R

Duodenal hormones reach the exocrine


pancreas via general circulation
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 37

Secretin similar to gastrin, with the 5 amino acids at the carboxyl


Secretin is produced by S cells in the duodenum and terminus identical to both hormones. CCK is also sul-
jejunum. Similar to gastrin, secretin is produced by fated on a tyrosine that is the seventh amino acid from
posttranslational processing of a larger preprosecretin the carboxy terminus. CCK binds primarily to the
molecule. Most secretin is a carboxyl amidated CCK1 receptor (formerly called the CCKA receptor),
27-amino acid peptide. whereas gastrin preferentially binds to the CCK2 recep-
The primary stimulus for secretin release is a tor. Both hormones can weakly interact with the other’s
decrease in duodenal pH. The threshold pH value for receptor, and desulfation of CCK increases its affinity
secretin release is 4.5. Circulating secretin levels increase for the CCK2 receptor. The CCK1 receptor is linked
rapidly (approximately 10 minutes) after acidified to protein kinase C–dependent and Ca2 þ-dependent
chyme passes through the pyloric sphincter into the du- pathways.
odenum. The exact mechanism by which Hþ induces The primary stimulus for CCK secretion is the pres-
secretin release from S cells is unclear. There is evidence ence of long-chain fatty acids or monoglycerides in
for a direct action of Hþ on S cells as well as evidence for the small intestine (see Fig. 2-8). CCK secretion is also
indirect actions through enteric neurons and through a induced by glycine-containing dipeptides and tripep-
phospholipase A2–like secretin-releasing factor. tides. The mechanism by which any of these act to
The primary short-term action of secretin is the stimulate CCK release is obscure, although there is
stimulation of the secretion of a bicarbonate-rich some evidence for a postabsorptive effect of lipids after
fluid from the pancreatic and biliary ducts during their assembly into chylomicrons. There is also evi-
the intestinal phase of the digestive period (see dence for a CCK-releasing peptide (CCK-RP) that is
Fig. 2-8). Secretin acts through the secretin receptor, released luminally from enterocytes and stimulates
which is linked to cAMP-dependent pathways. Signal- CCK release through binding to a CCK-RP receptor
ing from the secretin receptor opens apical Cl chan- on the apical membrane of I cells. Like secretin,
nels (cystic fibrosis transmembrane conductance CCK primarily regulates pancreatic and biliary
regulator or CFTR) thereby increases the flow of function. In the pancreas, CCK stimulates enzyme
transport Cl (and, through paracellular osmotic secretion from the acinar cells (see Fig. 2-8). The
drag, water) into the lumen. Cl is then exchanged CCK1 receptor increases intracellular DAG and Ca2 þ,
for HCO3. Upregulation of this process by secretin which results in the exocytosis of enzyme-containing
can occur through the opening of preexisting CFTR secretion granules. CCK also has a permissive effect
transporters in the apical membrane and through the on the ability of secretin to stimulate bicarbonate
exocytotic insertion of transporter-containing vesicles secretion.
into the membrane. The importance of the CFTR chan- CCK is a strong stimulator of gallbladder con-
nel to pancreatic function underlies the dysfunction of traction, and CCK deficiency disorders have been
pancreatic secretion observed in patients with cystic linked to impairment of gallbladder contraction
fibrosis. and cholelithiasis (gallstones). CCK induces gall-
Secretin also binds to its receptor on the pancreatic bladder contraction both directly and indirectly
acinar cells. Although secretin has a minimal effect on through activation of vagal afferent neurons. CCK
acinar cells by itself, secretin synergizes with the hor- also stimulates bile secretion into the duodenum
mone CCK to further enhance pancreatic enzyme through promoting relaxation of the sphincter of
secretion over that achieved by CCK alone. Secretin the hepatopancreatic ampulla (sphincter of Oddi).
may also function as an enterogastrone by inhibiting This latter action on hepatobiliary function is likely
stomach acid secretion. due to the CCK-dependent release of inhibitory
neurotransmitters, such as nitric oxide, from enteric
neurons. As mentioned, CCK also inhibits gastric
Cholecystokinin emptying, which reduces duodenal acidity and
CCK is a 33-amino acid peptide produced by the I cells allows emulsification, digestion, and absorption of
of the duodenum and jejunum. CCK is structurally lipids.
38 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

Motilin and Stimulation of Gastric and and glucagon-like peptide-1 (GLP-1). These peptides
Small Intestinal Contractions During the (or analogs thereof) are currently being investigated
Interdigestive Period for the treatment of type 2 diabetes mellitus (see
Motilin is a 22-amino acid peptide produced from a Chapter 3). An important feature of incretins is that their
114-amino acid prepromotilin and secreted by the ability to increase insulin secretion is strongly dependent
M cells of the small intestine. Motilin secretion is on glucose levels. This means that incretin analogs pose a
inhibited by the presence of food or acid in the small low risk for inducing severe hypoglycemia (low blood
intestine and is stimulated by alkalinization of the sugar) because once blood glucose falls, the effect of
small intestine. incretins is terminated.
Circulating motilin levels peak every 1 to 2 hours In general, GIP and GLP-1 act through Gs-coupled
in fasting individuals, in phase with the MMC. The receptors on b cells, which increase cAMP. This acts in
MMC is a set of organized contractions that move a permissive or synergistic manner with the main glu-
aborally from the stomach to the ileum and clean cose/adenosine triphosphate (ATP)-dependent path-
the stomach and small intestines of indigestible way that leads to an increased intracellular Ca2 þ and
particles. The MMC may also prevent the colonic the release of insulin. For example, cAMP-EPAC sig-
bacteria from migrating into the small intestine. naling (see Chapter 1) may promote the docking
Motilin may function to either initiate or integrate and regulated exocytosis of secretory vesicles in b cells.
the MMC. Incretins also enhance the synthesis of insulin and
The motilin receptor is a GPCR that activates the of proteins that sensitize the b cells to glucose levels,
phospholipase C signaling pathway. The motilin recep- such as the glucose transporter, GLUT-2, and
tor also binds and is activated by the macrolide antibi- hexokinase.
otic, erythromycin (see Table 2-2). Erythromycin and
other motilin receptor agonists are used in the treat-
Gastric Inhibitory Peptide/Glucose-
ment of delayed gastric emptying (gastroparesis),
Dependent Insulinotropic Peptide
which is common in patients with diabetes mellitus
and in some postsurgical patients. GIP is a 42-amino acid peptide secreted by the K cells
of the small intestine and is a member of the secretin
gene family. The primary stimulus for GIP release is
the presence of long-chain fatty acids, triglycerides,
INSULINOTROPIC ACTIONS OF glucose, and amino acids in the lumen of the small
GASTROINTESTINAL PEPTIDES intestine.
(INCRETIN ACTION) GIP was first discovered as an enterogastrone in an-
Elevated circulating levels of nutrients, particularly imal models, in which it inhibited gastric acid secre-
blood glucose, are strong stimuli of insulin secretion tion and intestinal motility. However, physiologic
from the pancreatic b cells (see Chapter 3). The pos- levels of GIP have only a modest effect on stomach
sibility that GI hormones also regulate the secretion function in humans. In contrast, GIP has an important
of insulin was revealed by observations that oral ad- physiologic role as an incretin. GIP knockout mice dis-
ministration of glucose caused a greater rise in insulin play a reduced ability to maintain normal blood glu-
than did glucose administered by an intravenous cose levels after an oral glucose load (impaired glucose
route. This enteroinsular response gave rise to the tolerance).
concept of incretins. In this model, an enteroendo- In rare cases, the GIP receptor is inappropriately
crine cell type senses nutrients in the GI tract and expressed on cells of the zona fasciculata of the adrenal
releases a hormone (an incretin), which, in turn, prepares cortex (see Chapter 7). These patients display enlarged
the pancreatic b cells for the impending rise in blood nu- adrenals and food-induced hypercortisolism. In these
trients (primarily blood glucose). There are two incretins patients, food in the small intestine stimulates the re-
in humans, gastric inhibitory peptide (GIP; also re- lease of GIP, which then stimulates cortisol production
ferred to as glucose-dependent insulinotropic peptide), by the adrenal cortex (see Chapter 7).
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 39

Glucagon-like Peptide-1 inhibitors of enzymatic degradation are currently


The glucagon gene is an example of a gene that en- under investigation for enhancing pancreatic b-cell
codes a large precursor protein (preproglucagon), function in type 2 diabetic patients.
which is proteolytically processed to form active and
inactive peptides (Fig. 2-9). Furthermore, the prohor-
mone convertases that digest preproglucagon display
ENTEROTROPIC ACTIONS OF
cell-specific expression, so different products are re-
GASTROINTESTINAL HORMONES
leased from different cell types. In the a cells of the An important characteristic of many hormones is their
endocrine pancreas, the active product is glucagon ability to promote the growth of their target tissues.
(see Chapter 3). In contrast, intestinal L cells express This tropic effect helps to maintain the health and in-
preproglucagon but secrete GLP-1 and GLP-2 as bio- tegrity of the target tissues and optimizes the ability of
logically active peptides. GLP-1 is stimulated by the target tissues to perform their differentiated functions.
presence of free fatty acids and glucose in the lumen In addition to the actions of GI hormones on the
of the ileum and colon. GLP-1 secretion is also in- maintenance of healthy GI structure and physiology,
creased by neuronal pathways stimulated by free fatty the tropic actions of GI hormones are of current
acids and glucose in the upper small intestine. GLP-1 is clinical interest for several reasons, including the
co-secreted with the other glucagon-derived peptide, following:
GLP-2, and peptide YY (which is not structurally re-
lated to glucagon). The tropic effect of GLP-2 is dis- n The promotion of hypertrophy and hyperplasia of
cussed later. GI tissues, which sometimes progress to cancer, by
Like GIP, GLP-1 acts as an incretin. GLP-1 knock- the excessive secretion of a GI hormone (usually
out mice have impaired glucose tolerance. GLP-1, from a tumor)
along with peptide YY, also appears to be a component n The ability of the GI tract to adapt to a diseased por-
of the ileal brake, in which free fatty acids and carbo- tion of the tract, and/or corrective surgery that in-
hydrates in the ileum inhibit gastric emptying through volves resection or bypass of a GI segment
increased secretion of GLP-1 and peptide YY. This en- n The ability to grow new pieces of GI tissue in vitro
terogastrone action of GLP-1 further enhances the (i.e., tissue-engineered neointestine) from pluripo-
ability of the organism to control excessive blood glu- tential or stem cells, which can be used for replace-
cose excursions. A problem with the therapeutic use ment of diseased or resected portions
of native GLP-1 is the fact that it is rapidly degraded. n The ability to promote pancreatic islet growth and
The use of more stable analogs, called exendins, and neogenesis in diabetic patients

Preproglucagon

SP GRPP GLUC GLP-1 GLP-2

Products from FIGURE 2-9 n Cell-specific processing of


GRPP GLUC GLP-1 GLP-2
pancreatic A cell preproglucagon.
Inactive Active Inactive

Products from
GRPP GLUC GLP-1 GLP-2
intestinal L cell

“Glicentin”-inactive Active Active


40 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

Gastrin receptor function may shift the cell toward net


In addition to its well-established role in the regula- proliferation.
tion of gastric acid secretion, gastrin exerts several
other effects on the stomach and GI tract. The second Glucagon-like Peptide-1
most important action of gastrin is its developmental One of the most exciting and promising aspects of
and trophic effect on the gastric mucosa. Gastrin enterotropic actions of GI hormones is the tropic
knockout mice display poorly differentiated gastric effect that GLP-1 has on pancreatic islet develop-
mucosa, with a reduced number of ECL and parietal ment and growth, particularly with respect to the
cells. In contrast, patients suffering from Zollinger- b cells. GLP-1 has been shown to induce differentiation
Ellison syndrome (see earlier) exhibit hypertrophy of human islet stem cells into b cells in vitro. In mice
and hyperplasia of the gastric mucosa, as well as en- and rats, GLP-1 and exendin-4 have protected against
larged submucosal rugal folds. Overgrowth is particu- surgically and chemically induced diabetes, increased
larly true for the ECL cell population. Although ECL b-cell mass and neogenesis, and inhibited b-cell apop-
cell proliferation can progress to carcinoid tumor tosis. Further, GLP-1 receptor knockout mice do not
formation, this is rare and usually requires other display exendin-4-induced regeneration of islets after
abnormalities. As discussed earlier, progastrin and partial pancreatectomy. Thus, GLP-1 or analogs may
glycine-extended gastrin (G-Gly) appear to promote become valuable reagents in the treatment of diabetic
the proliferation of colonic mucosa. patients whose b-cell mass has been compromised.
Gastric acid, through its effects on D cells and
somatostatin release, inhibits the growth of G cells.
Glucagon-like Peptide-2
Thus, long-term inhibition of gastric acid production
(e.g., with pharmacologic proton pump inhibitors or GLP-2 is co-secreted with GLP-1 by the intestinal L
H2 receptor blockers) can lead to an overgrowth of cells. Unlike GLP-1, GLP-2 does not have an insulino-
antral G cells. tropic action. GLP-2 binds to its own receptor (the
GLP-2 receptor) and has potent trophic effects on
the intestines. In fact, evidence of this effect was first
Secretin and Cholecystokinin discovered in a patient who presented with a massive
CCK has a direct effect on pancreatic acinar cells overgrowth of the small intestine. The patient was also
that promotes their maintenance and growth. found to have a tumor in the kidney that was produc-
Secretin inhibits pancreatic ductal cell growth ing large amounts of glucagon-related peptides. GLP-2
through binding to the secretin receptor. In contrast, has been used to prevent mucosal atrophy in patients
the secretin-related neurotransmitter, VIP, stimu- receiving total parenteral nutrition, and it promotes
lates ductal growth through the VIP receptor (called intestinal growth and adaptation in patients un-
VPAC1 receptor). In some ductal pancreatic adeno- dergoing resection of bowel. GLP-2 also has positive
carcinomas, the secretin receptor is defective, but effects on hexose transport and may enhance other
the VPAC1 receptor is intact. Thus, loss of secretin absorptive functions of intestinal villi.
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 41

S U M M A R Y

1. Gastrointestinal (GI) hormones are produced by 4. Motilin is secreted by the M cells of the small intestine
enteroendocrine cells. GI hormones are peptides during the interdigestive phase (i.e., in between
or proteins and bind to G-protein-coupled recep- meals), in phase with the migrating myoelectric com-
tors on their target cells. GI hormones are pro- plex. Motilin promotes emptying of the stomach and
duced by specific cell types that reside in specific small intestines. The motilin receptor is activated by
regions of the GI tract. The secretion of GI hor- erythromycin, which can be used to treat delayed gas-
mones is stimulated primarily by luminal secreto- tric emptying (gastroparesis).
gogues and by neuronal (enteric and autonomic) 5. GI hormones called incretins are secreted in
and paracrine signals. response to luminal nutrients (especially glucose)
2. Gastrin plays a major role in the stimulation of gas- and increase the ability of blood glucose to stimulate
tric acid secretion. Gastrin is secreted by G cells in insulin secretion from the pancreatic islets of Lan-
the stomach antrum in response to amino acids and gerhans. Incretins include gastric inhibitory peptide
peptides in the antral lumen and in response to neu- (GIP), which has been named more recently for its
ronal stimulation. The primary secreted form of gas- incretin effect as glucose-dependent insulinotropic
trin by the stomach is the 17-amino acid G-17 form. peptide. GIP is secreted from the K cells of the small
G-17 has a cyclized glutaminyl residue at its N- intestine. Another important incretin is glucagon-
terminus and an amidated glycine at its C-terminus, like peptide-1 (GLP-1), which is secreted by the in-
which increase the biologic half-life of secreted gastrin. testinal L cells. Because of their ability to sensitize
Gastrin binds to the CCK2 receptor and acts primarily insulin-producing b cells to glucose, incretins are
by stimulating enterochromaffin-like cells (ECL cells) being tested for the treatment of type 2 diabetes mel-
to secrete histamine. Histamine then stimulates the litus (T2DM; see Chapter 3).
parietal cells of the stomach to secrete HCl. 6. GI hormones also have important trophic effects.
3. The major enteroendocrine cells of the duodenum Gastrin stimulates the growth of the gastric
and jejunum are the S cells and I cells, which secrete mucosa, especially the ECL cells and submucosa.
secretin and cholecystokinin (CCK), respectively. Secretin and CCK promote the growth of exocrine
Secretin is released primarily during the intestinal pancreas tissue. GLP-1 promotes b-cell prolifera-
phase of a meal in response to increased acidity in tion, which may prove an important function of
the duodenum. Secretin promotes the secretion of GLP-1 in the treatment of T2DM. GLP-2, which
a bicarbonate-rich fluid from the bile duct and pan- is related to, but is a separate hormone from,
creatic ducts, which empty into the duodenum. GLP-1, promotes GI mucosal growth and is used
CCK promotes the contraction of the gallbladder to treat patients at risk for GI mucosal atrophy.
and relaxation of the sphincter of the hepatopan- 7. Zollinger-Ellinger syndrome is caused by a gastrin-
creatic ampulla, thus promoting the emptying of producing tumor. Patients have ulcerations of the
bile into the duodenum. CCK also stimulates esophagus, stomach, and duodenum and overgrowth
enzyme secretion from pancreatic acinar cells. of the stomach mucosa and rugal submucosal folds.
42 ENDOCRINE AND REPRODUCTIVE PHYSIOLOGY

3. What is the relation between gastric emptying and


SELF-STUDY PROBLEMS
gastrin secretion from duodenal S and I cells?
4. What are the effects of CCK on the following?
1. What are the three phases of the digestive period? a. Pancreatic bicarbonate secretion
Which one has the greatest release of gastrin? Why? b. Pancreatic enzyme secretion
2. When administered during the interdigestive c. Biliary bicarbonate secretion
period, what are the predicted effects on gastrin d. Contraction of the gallbladder muscularis
secretion of the following experimental agents? e. Contraction of the sphincter of Oddi
a. A somatostatin antagonist 5. What is the relation between GLP-1 and glucagon?
b. A mix of amino acids in the antral lumen 6. Define incretin. Name two incretins.
c. Increased acidity in the antral lumen 7. What enterotropic effect is observed in patients
d. A muscarinic agonist with Zollinger-Ellison syndrome?
e. Gastrin-releasing peptide 8. Why does erythromycin promote gastric emptying?

KEYWORDS AND CONCEPTS SUGGESTED READINGS


Fourmy D, Gigoux V, Reubi JC: Gastrin in gastrointestinal diseases,
n Acetylcholine Gastroenterology 141:814–818, e811–e813, 2011.
n Amidated gastrins Ishii H, Sato Y, Takei M, et al: Glucose-incretin interaction revisited,
n Autonomic nervous system Endocr J 58:519–525, 2011.
Poitras P, Peeters TL: Motilin, Curr Opin Endocrinol Diabetes Obes
15:54–57, 2008.
Rehfeld JF: Incretin physiology beyond glucagon-like peptide 1 and
For full list of keywords and concepts see Student glucose-dependent insulinotropic polypeptide: Cholecystokinin
Consult and gastrin peptides, Acta Physiol 201:405–411, 2011.
ENDOCRINE FUNCTION OF THE GASTROINTESTINAL TRACT 42.e1

n G-protein-coupled receptors
KEYWORDS AND CONCEPTS
n Growth hormone secretogogue
n Acetylcholine n HCl
n Amidated gastrins n Hormone
n Autonomic nervous system n I cells
n CCK n Impaired glucose tolerance
n CCK1 receptor n Incretins
n Cephalic phase n Incretion action
n Chief cells n Intestinal phase
n Cholecystokinin (CCK) n Intrinsic factor
n Chyme n Intrinsic regulators
n Duodenum n Migrating myoelectric complex (MMC)
n Endocrine glands n Motilin
n Enteric nervous system n Mucigens
n Enteroendocrine cell n Oxyntic cells
n Enterogastrones n Paracrine
n Enterochromaffin-like (ECL) cells n Parietal cells
n Enterotropic action n Pentagastrin
n Erythromycin n Peptide YY
n Exendins n Preprogastrin
n Exocrine pancreas n Pyloric antrum
n Extrinsic regulators n S cells
n Food-induced hypercortisolism n Secretin
n Fundus and body n Secretin-releasing factor
n G-17 n Secretogogues
n Gallbladder n Somatostatin
n Gastric phase n Stomach
n Gastrin n Vagal parasympathetic nervous system
n Gastrin-releasing peptide (GRP) n Vagovagal reflex
n Gastroparesis n Vasoactive intestinal peptide
n Ghrelin n Vitamin B12
n Glucose-dependent/insulinotropic peptide n Zollinger-Ellison syndrome

Das könnte Ihnen auch gefallen