You are on page 1of 10

Endocrinology, 2021, Vol. 162, No.

7, 1–10
doi:10.1210/endocr/bqab065
Mini-Review

Mini-Review

The Role of Incretins on Insulin Function and


Glucose Homeostasis

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


Jens Juul Holst,1 Lærke Smidt Gasbjerg,1 and Mette Marie Rosenkilde1
1
Department of Biomedical Sciences and the NovoNordisk Center for Basic Metabolic Research, Faculty
of Health Sciences, University of Copenhagen, The Panum Institute, Copenhagen N, DK-2200 Denmark
ORCiD numbers: 0000-0001-6853-3805 (J. J. Holst); 0000-0002-7880-8515 (L. S. Gasbjerg); 0000-0001-9600-3254 (M. M.
Rosenkilde).

Abbreviations: Ex-9, exendin 9-39; GIGD, gastrointestinally mediated glucose disposal; GIP, glucose-dependent insulinotropic
polypeptide; GLP-1, glucagon-like peptide-1; IV, intravenous
Received: 26 February 2021; Editorial Decision: 22 March 2021; First Published Online: 30 March 2021; Corrected and Typeset:
1 June 2021.

Abstract
The incretin effect—the amplification of insulin secretion after oral vs intravenous ad-
ministration of glucose as a mean to improve glucose tolerance—was suspected even
before insulin was discovered, and today we know that the effect is due to the secretion
of 2 insulinotropic peptides, glucose-dependent insulinotropic polypeptide (GIP) and
glucagon-like peptide-1 (GLP-1). But how important is it? Physiological experiments have
shown that, because of the incretin effect, we can ingest increasing amounts of amounts
of glucose (carbohydrates) without increasing postprandial glucose excursions, which
otherwise might have severe consequences. The mechanism behind this is incretin-
stimulated insulin secretion. The availability of antagonists for GLP-1 and most recently
also for GIP has made it possible to directly estimate the individual contributions to post-
prandial insulin secretion of a) glucose itself: 26%; b) GIP: 45%; and c) GLP-1: 29%. Thus,
in healthy individuals, GIP is the champion. When the action of both incretins is pre-
vented, glucose tolerance is pathologically impaired. Thus, after 100 years of research,
we now know that insulinotropic hormones from the gut are indispensable for normal
glucose tolerance. The loss of the incretin effect in type 2 diabetes, therefore, contributes
greatly to the impaired postprandial glucose control.
Key Words: glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), hormone antagon-
ists, hormone coagonists, exendin 9-39

Bayliss and Starling, who in 1902 discovered secretin (1), and other scientists investigated the hypoglycemic effects
the very first hormone, were convinced that not only the of intestinal extracts and secretin preparations. Thus, the
exocrine, but also the endocrine secretion from the pan- incretin concept, the idea that the gut regulates glucose
creas, discovered by von Mering and Minkowski in 1889 levels via effects on the endocrine pancreas, is older than
(2), was regulated by intestinal factors, and both they insulin itself. Although Zunz and La Barre, like Bayliss

ISSN Online 1945-7170


© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-
NoDerivs licence (http://creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial reproduction https://academic.oup.com/endo   1
and distribution of the work, in any medium, provided the original work is not altered or transformed in any way, and
that the work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
2 Endocrinology, 2021, Vol. 162, No. 7

and Starling, were convinced that secretin was the most However, in animal experiments, immunoneutralization
important hypoglycemic substance (3), they nevertheless of GIP with potent antiserum samples did not eliminate
tried to distinguish between “excretins” (stimulating the the incretin effect (14), and in people with resections of
exocrine secretion from the pancreas) and “incretins” with different parts of the small intestine, there was no correl-
predominant hypoglycemic effects. In those days, the physi- ation between the incretin effect and the GIP responses
ologists made many impressive and daring (and ethically (15). Clearly, something was missing. The missing hormone
problematic) experiments such as anastomosing a pancre- was glucagon-like peptide-1 (16), a product of intestinal
atic vein from a donor dog to the jugular vein of a recipient expression of proglucagon, the precursor of the pancre-
dog, to demonstrate that blood glucose falls in the latter atic hormone glucagon. The gene encoding proglucagon is
after injection of a secretin preparation into the donor dog, also expressed in the so-called L cells, endocrine cells of
but it was only after the development of the radioimmuno- the intestinal epithelium (17), where the precursor is pro-

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


assay for insulin (4) that it became possible to quantify cessed to release 2 glucagon-containing peptides, glicentin
and determine the importance of the incretin effect for in- and oxyntomodulin, and 2 glucagon-like peptides, GLP-1
sulin secretion. In 1964, McIntyre and colleagues (5) re- and GLP-2 (18, 19). Of these products, oxyntomodulin and
ported that intestinal administration of glucose elicited a GLP-1 both are insulinotropic, but whereas the concentra-
much greater insulin response than intravenous (IV) in- tions of oxyntomodulin normally are too low to stimulate
jections and they correctly deduced that intestinal factors, insulin secretion (20), the 100-fold more potent GLP-1 pro-
incretins, were responsible. In 1971, Brown and Dryburgh vides a very powerful stimulus (21) and in mimicry experi-
had sequenced a new peptide, gastric inhibitory polypep- ments clearly qualified as a new incretin; actually, at first
tide, GIP, isolated from impurities of cholecystokinin on sight, more potent than GIP (22). Today, there is agreement
the basis of its inhibitory effects on acid secretion from that GLP-1 and GIP are the most important incretin hor-
canine gastric pouches (6), but in 1973 Dupre and Brown mones, and it was determined early on that in mice with
identified the insulinotropic effects of GIP (7) (which, be- deletions of the receptors for both GIP and GLP-1, very
cause the inhibitory effects on gastric secretion turned out little incretin effect remained (23). However, it is also clear
not to be physiologically relevant, was renamed “glucose- that these mouse experiments do not tell us much about the
dependent insulinotropic polypeptide,” retaining the nice importance of the incretin effect. After oral glucose gavage,
acronym, GIP). Many subsequent studies confirmed its there are observable differences between the glucose excur-
insulinotropic actions, and in 1989, Nauck et al could sions in animals with and without receptor deletions, but
show in mimicry experiments that GIP fulfilled rigorous they are small and barely significant. In addition, it is pos-
criteria for being a physiologically important incretin hor- sible that rodents, not the least mice, depend less on the
mone (8). Thus, GIP, infused intravenously in amounts that incretin effect in their regulation of glucose homeostasis
copied its plasma concentrations after oral glucose, had ef- than humans and more on neural regulation (24, 25), and
fects on insulin secretion that were similar to those elicited it is therefore questionable how much these experiments
by the oral glucose. can tell us about human conditions.
But what about secretin? Secretin had been isolated and
sequenced in Stockholm in 1970 by Mutt and colleagues
(from extracts of intestines from 10 000 hogs) (9), and it The Importance of the Incretin Effect
was now possible to synthesize the peptide and develop So, how important is the incretin effect in people? This
radioimmunoassays. Infusion experiments clearly con- is really an important question, because one of the char-
firmed the old observations that secretin would stimulate acteristics of type 2 diabetes is a more or less complete
insulin secretion (actually even in people with type 2 dia- loss of the incretin effect (13). This raises the ques-
betes [10]), but the circulating concentrations of secretin tion whether type 2 diabetes is an incretin disease, al-
were extremely low (1-2 pmol/L) and even after maximal though the loss of the incretin effect does not usually
stimulation with intraduodenal acid, they did not reach top the list of pathogenic factors. The importance of the
levels sufficient to stimulate insulin secretion (11). But incretin effect in healthy individuals can be illustrated
there had to be other incretin(s). With the insulin radio- in experiments in which the so-called gastrointestinally
immunoassay, it was possible to quantify accurately the mediated glucose disposal (GIGD [26]) is determined.
incretin effect by comparing insulin responses to IV and The approach is simple: One measures the total amount
oral administrations of glucose given in amounts that re- of IV glucose that is required to mimic the plasma ex-
sulted in identical glucose concentrations (isoglycemia), cursions after a given oral dose and compares the two.
and it could be calculated that up to 70% of the response Using this technique, Nauck et al (27) first showed
to oral glucose could be due to the incretin effect (12, 13). that GIGD strongly depends on the amount of glucose
Endocrinology, 2021, Vol. 162, No. 7 3

ingested. Thus, with 25-g glucose given orally, an infu- some early studies (29); it was shown that a 25-g dose
sion of about 20 g of glucose was required to copy the of glucose resulted in almost the same insulin secretion
plasma glucose response. However, with 50 g of glucose, (measured as C-peptide) whether it was given intraven-
it also took about 20 g to copy the curve, and with 75 g ously or orally, and it was concluded that the “incretin
it also took 20 g to copy the curve. It follows that the effect” was a hoax. But we now know that a) a dose of
plasma excursions after the 3 doses of oral glucose were glucose as small as 25 g elicits a GIGD of only 20% (be-
virtually identical. In other words, the body is capable of cause it took 20 g of IV glucose to mimic the 25-g oral
disposing of orally ingested glucose so rapidly and effi- dose—see earlier), and b) IV administration gives a much
ciently that the plasma glucose excursions remain con- larger plasma glucose response than oral dosing, ruining
stant and low in spite of widely different amounts taken the principle of isoglycemia, and therefore the possibility
in. Thus, this gastrointestinal mechanism is capable of to gauge the incretin response properly.

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


removing up to three-quarters of the absorbed glucose The incretin hormone responses were also deter-
from the circulation. This shows how efficiently we nor- mined in the cited dose-response experiments (Fig. 1) and
mally regulate our postprandial glucose concentrations. show some interesting features (27, 28). GLP-1 secretion
But what is the mechanism behind this remarkable glu- showed a dose response relationship that corresponded
cose disposal? it turns out to be insulin secretion, which more or less with the insulin responses. GIP secretion
increases progressively with increasing glucose doses was a bit different, showing an early rise to a peak value,
(27). In other words, the incretin effect secures normal which was more or less the same with all doses, and a
and healthy postprandial glucose excursions almost no subsequent plateau, the duration of which was propor-
matter how large the dosing is (125 g has also been in- tional to the glucose dose. This profile probably reflects
vestigated with similar results [28]). Funnily enough, the the gastric emptying of glucose, which is regulated so as
importance of the incretin effect was nearly missed in to ensure a relatively constant emptying rate of nutrients,

Figure 1. Localization of glucose-dependent insulinotropic polypeptide (GIP)- and glucagon-like peptide-1 (GLP-1)-secreting cells in the gastrointes-
tinal tract (left) and secretion profiles of GIP, GLP-1, and insulin in response to ingestion of low (25 g), medium (50-75 g) or high (100-125 g) amounts
of glucose (right). Curves adapted from Nauck et al (27) and Bagger et al (28).
4 Endocrinology, 2021, Vol. 162, No. 7

corresponding, in the case of glucose, to about 4 kcal/min diabetes rather than being a primary deficiency (38). More
(30, 31). Since this translates into a more or less constant about this later.
exposure to glucose of the duodenal mucosa, from where The evidence regarding the importance of the incretin
most of the GIP is secreted, an enhanced secretion of GIP hormones for glucose tolerance presented so far is based on
will be maintained as long as there is glucose left in the the principles of the mimicry experiments: If we mimic the
stomach. GLP-1 secretion occurs from slightly more distal plasma profiles observed in vivo with infusions of glucose/
gut segments, which means that most of the L cells are hormones, do we get the same responses? Although such
likely to escape stimulation because of early absorption experiments represent necessary conditions for conclu-
of glucose. Actually, the grossly exaggerated GLP-1 re- sions on causality, they are not sufficient. Other hormones
sponses to more distal glucose exposure in patients with could easily be involved and neural, renal, hepatic and, in
gastric bypass operations (32) is another illustration of fact, gut mechanisms could be overlooked. The double-

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


this. Both the L cells and the GIP-secreting K cells ex- knockout experiments referred to previously could be con-
press the sodium-coupled glucose transporter, SGLT1, sidered sufficient evidence (with the trivial reservation that
and their secretory responses can be demonstrated to de- the receptors were truly knocked out and that knockout
pend on this transportation. In this way, the secretion of did not result in compensatory adaptations, which is more
the incretin hormones becomes proportional to the rate of likely than not), but were not carried out in the relevant
glucose absorption in the gut segment, where this occurs. species. To solve the problem, the application of hormone
(33, 34). receptor antagonists would be a useful approach. Such
The conclusion is that normal postprandial glucose tol- antagonists must be acceptable for human use, which is a
erance to a very large extent is determined by the incretin major hindrance, but this has recently become possible for
effect, which in turn is due to the secretion of the incretin both hormones (Fig. 2).
hormones. Thus, the stimulatory effect of glucose alone
on insulin secretion is far from sufficient. As briefly men-
tioned earlier, the incretin effect is very much reduced or Glucagon-Like Peptide-1 Receptor
frankly missing in patients with type 2 diabetes, and one Antagonism
may ask why that is. When it became possible to measure In 1992, Eng et al isolated a 39 amino acid peptide,
the plasma concentrations of GIP and GLP-1, it was nat- exendin-4, from the saliva of the Gila monster (Heloderma
ural to look for secretion of these hormones in diabetes. suspectum) (39), which was capable of activating receptors
There are strong indications that the postprandial secretion om pancreatic acinar cells, but also found that a truncated
of GLP-1 is impaired in type 2 diabetes (35); perhaps not version, exendin 9-39 (40), could antagonize the actions
the early phase of meal responses (36), but particularly the of exendin-4 (39). Very soon after its discovery, exendin-4
late phase of the response, and the impairment is probably was demonstrate to be a full and potent agonist for the
more related to the negative influence of obesity than it is GLP-1 receptor, which had just been cloned, and the trun-
related to diabetes. However, it is also clear that impaired cated form, exendin 9-39 (Ex-9), was demonstrated to an-
secretion is not the most important explanation for the tagonize the actions of exendin-4 as well as GLP-1 on the
decreased incretin effect. Rather, the insulinotropic action GLP-1 receptor (41) (see Fig. 2). Interestingly, exendin-4,
of both hormones is impaired (37). The impaired incretin which has about 50% sequence homology with mamma-
effect is a very early event in type 2 diabetes, but it is the lian GLP-1 (regarding the first 30 amino acids) is not the
prevailing concept that the defect develops in response to GLP-1 of the Gila monster—this poisonous lizard has its

Figure 2. Central events in the history of the 2 incretin receptor antagonists, exendin(9-39)NH2 and GIP(3-30)NH2.
Endocrinology, 2021, Vol. 162, No. 7 5

own GLP-1 (which is much closer to mammalian GLP-1)


(42). Back to the antagonist: Two laboratories soon car-
ried out infusion experiments in humans of Ex-9 to elu-
cidate the role of the endogenous GLP-1 by blocking its
receptor (43, 44). Regarding safety, these experiments
went well, which is ironic considering that the use of syn-
thetic exendin-4 (exenatide) for diabetes therapy was
subsequently suspected of causing acute pancreatitis (45)—
leading a Food and Drug Administration officer to remark
in the New England Journal of Medicine (46) that this was
an expected result considering the toxicity of being bitten

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


by a Gila monster (the GLP-1 receptor agonists, including
exendin-4, were later completely acquitted with respect to
the pancreatitis suspicion [47]). Since then numerous ex-
Figure 3. Exendin(9-39)NH2 actions on pancreatic δ (delta), α (alpha),
periments have been carried out with Ex-9. Indeed, it is cur-
and β (beta) cells, which can result in increased insulin secretion.
rently being developed as a therapeutic agent (avexitide) to Glucagon-like peptide-1 (GLP-1) receptors are red, glucagon recep-
prevent reactive postprandial hypoglycemia after bariatric tors are blue, and somatostatin receptors black. The yellow peptide
surgery, which appears to be due to exaggerated GLP-1 se- is exendin(9-39)NH2, GLP-1 is red, glucagon is blue, and somatostatin
black. Blockage of the GLP-1R on δ cells by exendin(9-39)NH2 prevents
cretion and similarly exaggerated insulin responses, suffi- somatostatin inhibition of α cells. Increasing glucagon secretion may
cient to cause temporary hypoglycemia (ClinicalTrials.gov stimulate insulin secretion via the glucagon receptor in the β cells.
identifier: NCT03373435). In fact, avexitide was granted Exendin(9-39)NH2 may also inhibit GLP-1 action on beta cells or act as
inverse agonist.
breakthrough therapy designation by the Food and Drug
Administration in 2019 for this indication (postbariatric
hypoglycemia). speculation: Glucose levels and glucagon concentration do
In human experiments involving intraduodenal glu- increase during Ex-9 administration. In previously diabetic
cose infusions, Ex-9 increases plasma glucose excursions, individuals who undergo gastric bypass surgery, Ex-9 in-
decreases insulin responses, and increases glucagon re- variably reduces meal-induced insulin responses and im-
sponses, which would be expected based on the actions pairs glucose tolerance, while increasing GLP-1 responses
of exogenous GLP-1 (43, 48). The results of Ex-9 infu- further (55); the GLP-1 increase seems to be due to inter-
sion in the fasting state and during simple oral glucose ference with another paracrine interaction, this time in the
administration are less easy to understand, however. The gut between L cells and somatostatin-producing δ cells or
infusions invariably elevate glucose concentrations and in- somatostatinergic neurons (56), which are known to be
crease glucagon concentrations (44), but insulin responses stimulated by the increased GLP-1 secretion. When this
are more variable, and decreases as well as increases have effect of GLP-1 is blocked by Ex-9, somatostatin release
been found. The increases in glucagon would be assumed falls and GLP-1 secretion increases.
to arise because of antagonism of GLP-1 receptors, but What happens to GIP secretion during Ex-9 infusion?
where? The α cells do not seem to express GLP-1 receptors The answer is: nothing (57). This tells us that the K cells
(very few at least), and most people agree that the inhib- are not subject to a similar somatostatinergic regulation as
ition would be due to a relief of the paracrine inhibition the L cells, and that there is no “compensatory” increase in
within the islets by somatostatin produced by neighboring GIP secretion, when GLP-1 action is blocked.
δ cells, which express stimulatory GLP-1 receptors (49-
51). But in the fasting state there is not much GLP-1 se-
cretion. It has been shown that Ex-9 may act as an inverse Glucose-Dependent Insulinotropic
agonist (52, 53), inhibiting the activity of the receptor it Polypeptide Receptor Antagonism
binds to, and in that case it is conceivable that Ex-9 may Various methods for producing GIP antagonism have been
stimulate glucagon secretion by inhibiting the tonic activity implemented in animal experiments during previous dec-
of the δ cells (because of some constitutive activity of the ades, but only recently has it been possible to antagonize GIP
GLP-1 receptor) to which the α cells are coupled in a para- action in humans (see Fig. 2). In a systematic search for an-
crine feedback loop (54). Stimulated insulin secretion after tagonistic effects of truncations and substitutions of the GIP
Ex-9 may then be due to a paracrine action of glucagon sequence, a peptide with a high affinity for the GIP receptors,
on the β cells via their glucagon receptors, potentiated which also turned out to be a potent competitive antagonist,
by the increasing glucose levels (Fig. 3). This is not pure was identified, namely GIP 3-30 NH2 (58, 59). A truncated
6 Endocrinology, 2021, Vol. 162, No. 7

infusion). However, when C-peptide responses (as a more


accurate measure of β-cell secretion) were adjusted for the
different glucose responses, a clear impairment of glucose-
induced insulin secretion became apparent, more with the
GIP antagonist GIP(3-30)NH2 than with Ex9, and again
roughly additively with the combination of the 2 antag-
onists. In an accompanying editorial, Nauck and Meier
calculated the individual contributions to the effects (63)
and concluded that the incretin effect, deduced from the
contributions of each of the components, was distributed
Figure 4. Contribution of gut-derived factors to postprandial insulin with 26% from glucose alone (similar to the value found

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


secretion, expressed in per cent of the total C-peptide secretion (area in traditional estimates of the incretin effect) and with 45%
under curve, 0-240 minutes) following ingestion of 75-g glucose orally from GIP and 29% from GLP-1. These figures were subse-
with or without incretin receptor antagonist infusions in healthy indi-
quently confirmed by calculations from the individual data
viduals. Calculations performed by Gasbjerg et al (64). GIP, glucose-
dependent insulinotropic polypeptide; GLP-1, glucagon-like peptide-1. (Fig. 4) (64). Thus, it may be concluded that in healthy in-
dividuals, GIP is the most important incretin, perhaps con-
form of GIP, GIP 1-30 NH2, which is a full and equipotent sistent with the proximal location of the K cells (duodenum
agonist of the GIP receptor, had been known to exist for and proximal jejunum) and their immediate dependence on
some time (60), and because GIP, like GLP-1, is substrate for the outflow of carbohydrate from the stomach, whereas the
the proteolytic activity of dipeptidyl peptidase-4 (DPP-4), an GLP-1 effect sets in when ingested glucose reaches a bit
N-terminally truncated form of this molecule, GIP 3-30NH2, farther down into the intestine. Indeed, after a gastric by-
was predicted to exist. GIP 1-30 NH2 is found in the cir- pass operation, as alluded to earlier, the nutritional load
culation in very low picomolar concentrations (57), so the is pushed farther down the small intestine, and the GLP-1
concentrations of endogenous GIP 3-30NH2 are likely to be responses are huge (typically 10-fold increased), whereas
similarly low. However, infused in sufficient quantities, the GIP responses are unchanged, and almost all the increased
antagonist dose-dependently blocks all the actions of GIP to insulin secretion disappears with Ex-9 (55). It should not
a very high degree (> 80%), be it on insulin secretion, stimu- be forgotten, as discussed previously, that Ex-9 is a difficult
lation of adipose tissue blood flow and triglyceride uptake, or tool to work with; therefore, the quantitative data obtained
inhibition of bone resorption (61, 62). Whereas Ex-9 works may be somewhat misleading.
equally well in experimental animals and humans, the GIP
system shows clear differences between species, necessitating
the use of species-specific agonists and antagonists in studies The Incretins and Type 2 Diabetes
of GIP receptor–mediated activities (59). As mentioned, patients with type 2 diabetes have an im-
paired or absent incretin effect, and in an experiment in
patients with rather long-standing diabetes, infusions of
Combinations of Glucose-Dependent the 2 hormones in amounts recapitulating normal meal
Insulinotropic Polypeptide and Glucagon- responses, there was absolutely no effect on insulin se-
Like Peptide-1 Receptor Antagonism cretion during the conditions of a 15-mmol/L hyper-
In a recent study (57), antagonists of GIP and GLP-1 were glycemic clamp (65). In agreement with these findings,
combined in an attempt to finally define the incretin actions infusion of the GIP antagonist in patients with type 2 dia-
of the 2 hormones during an oral glucose tolerance test in betes had no effect on postprandial glucose levels (66),
healthy volunteers. Indeed, and as expected, both of the ant- whereas Ex-9 slightly increases postprandial glucose (67).
agonists significantly impaired glucose tolerance, and when Likewise, whereas even very high rates of GIP infusion
given together their effects were additive, bringing the glu- had very little effects on insulin secretion and glucose
cose responses into the range of pathologically impaired turnover in additional individuals with type 2 diabetes
glucose tolerance (peak levels increasing by ~ 3 mmol/L and (68), supraphysiological GLP-1 infusions were capable of
2-hour values from 6.4 to 7.5 mmol/L), clearly confirming restoring insulin secretion levels to values similar to those
the importance of the incretin hormones for postprandial observed in healthy controls during glucose infusion
glucose tolerance. Their immediate effect on insulin secre- alone (whereas glucose alone was similarly ineffective
tion was difficult to gauge because of the increasing blood in the patients). The reason for this difference between
glucose concentrations (compared to the saline control the 2 incretin hormones is still unknown. In a careful
Endocrinology, 2021, Vol. 162, No. 7 7

dose-response study involving increasing infusion rates of strong antidiabetic properties in phase 2 studies, exceeding
GLP-1 on top of a step-wise clamping of plasma glucose those of an established, long-acting GlP-1 receptor agonist
at increasing levels, the slopes of the GLP-1 effects on (dulaglutide), and it was suggested that its effectiveness was
the β-cell responsiveness to glucose (the latter calculated due to a combination of the GIP and the GLP-1 effect (75).
from cross-correlations of insulin secretion rates and glu- The coagonist also has strong inhibitory effects on appetite
cose clamp levels) could be determined (69). This slope and food intake (and eventually body weight), something
was clearly impaired in patients compared to controls, that has never been observed in human studies with acute
even if adjusted to the estimated maximal β-cell perform- GIP administration (and it has not yet been excluded that
ance. This clearly illustrated the decreased responsiveness the effect could be due solely to GLP-1 receptor agonism).
of the diabetic β cells to GLP-1, but it also showed that In contrast, in other experimental studies, GIP antagon-
GLP-1, even at relatively low infusion rates, was capable ists lowered body weight, particularly in combination with

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


of restoring β-cell function to the same levels as those ob- GLP-1 (76). The surprising effects of the coagonism are
served in the controls given glucose alone (69). In other currently under intense investigation, and some studies in-
words, in patients with type 2 diabetes, supraphysiological dicate that differential internalization of the GIP and GLP-1
administration of GLP-1 may improve β-cell secretion to receptors after exposure to the coagonist may explain its
levels similar to those of healthy controls in response to unexpected effect (77). At any rate, the phase 3 studies with
glucose alone (whose responses to combinations of glu- tirzepatide (78) support its suitability for therapy of type
cose and GLP-1 would, of course, be much larger). This 2 diabetes (according to press releases from Eli Lilly), and
effect of GLP-1 forms the background for its therapeutic future studies will probably show whether GIP agonism or
use in patients with type 2 diabetes, in whom a GLP-1 antagonism is preferable for metabolic interventions.
infusion may actually restore fasting glucose levels to However, the incretin story presented here clearly shows
completely normal levels (70). It must be emphasized, that in healthy individuals the β cells do not work alone—for
however, that the β-cell responses of the individual pa- normal glucose tolerance, the gastrointestinal tract and the
tient with type 2 diabetes depend strongly on the residual incretin hormones are absolutely indispensable. Although
β-cell function, and in patients with very poor β-cell re- not expected originally, the incretin system also plays an im-
serve, the effect of GLP-1 may not be sufficient to nor- portant role in the regulation of food intake, and the most
malize glucose levels (71). Thus, in patients with fasting recent research has shown that GLP-1 receptor agonists
glucose levels between 7 and 10 mmol/L, a “standard” and surprisingly also GIP/GLP-1 receptor coagonists have
1.2-pmol/kg × min infusion of GLP-1 was capable of powerful effects on the treatment of not only diabetes but
completely normalizing glucose levels (72); in those with also obesity. The incretins certainly have come of age.
fasting glucose levels between 10 and 15 mmol/L, glucose
levels were much improved and reached 6 to 7 mmol/L
within 4 hours; and in those with fasting values greater Additional Information
than 15 mmol/L there was a decrease in glucose levels Correspondence: Jens Juul Holst, MD, University of Copen-
of up to 10 mmol/L, but in spite of extended infusion, hagen, Department of Biomedical Sciences, The Panum Institute,
levels did not fall below 8 mmol/L, indicating that even 3 Blegdamsvej, Copenhagen, DK-2200 Denmark. Email: jjholst@
the slightest meal stimulus would bring glucose levels up sund.ku.dk.
Disclosures: J.J.H. appears on advisory boards for NovoNordisk
again, so it would never be possible to cause a sufficient
and MSD, and J.J.H. and M.M.R. have collaborated with Lilly on
and lasting improvement in glycemic control in this way coagonist projects. J.J.H. and M.M.R. are founders of Antag Phar-
(72). In such a patient, a combination of basal insulin and maceuticals. L.S.G. has nothing to disclose.
GLP-1 works wonders (73). Data Availability: No new data have been included in this review.
As is evident from the process described earlier, the
focus has been on the development of GLP-1 analogues for
type 2 diabetes, whereas GIP was considered unattractive.
References
Some experiments indicated that improved glycemic con- 1. Mering Jv,Minkowski O. Diabetes mellitus nach Pankreasexstirpation.
trol in the patients might lead to recovery of some of the Archiv f experiment Pathol u Pharmakol. 1890;26:371-387.
2. von Mering J, Minkowski O. Diabetes mellitus and pancreas ex-
insulinotropic effects of GIP (74). But the truth is that
tirpation. Arch Exp Path Pharmak. 1889;21:371-390.
these improvements were very small and far from levels
3. Zunz E, La Barre J. Contributions a l’étude des variation
that would be therapeutically relevant. However, more re- physiologiques de la sécrétion interne de pancréas: relations
cently monomolecular GIP/GLP-1 coagonists have been entre les secretions externe et interne du pancréas. Archs Int
developed, and one of these, tirzepatide, showed very Physiol Biochim. 1929;31(1):20-44.
8 Endocrinology, 2021, Vol. 162, No. 7

4. Berson SA, Yalow RS. Quantitative aspects of the reaction 23. Hansotia T, Baggio LL, Delmeire D, et al. Double incretin re-
between insulin and insulin-binding antibody. J Clin Invest. ceptor knockout (DIRKO) mice reveal an essential role for the
1959;38(11):1996-2016. enteroinsular axis in transducing the glucoregulatory actions of
5. McIntyre N, Holdsworth CD, Turner DS. New interpretation of DPP-IV inhibitors. Diabetes. 2004;53(5):1326-1335.
oral glucose tolerance. Lancet. 1964;2(7349):20-21. 24. Pacini G, Thomaseth K, Ahrén B. Contribution to glucose toler-
6. Brown JC, Dryburgh JR. A gastric inhibitory polypep- ance of insulin-independent vs. insulin-dependent mechanisms
tide. II. The complete amino acid sequence. Can J Biochem. in mice. Am J Physiol Endocrinol Metab. 2001;281(4):E693
1971;49(8):867-872. -E703.
7. Dupre J, Ross SA, Watson D, Brown JC. Stimulation of in- 25. Ahrén B, Pacini G. Glucose effectiveness: lessons from studies
sulin secretion by gastric inhibitory polypeptide in man. J Clin on insulin-independent glucose clearance in mice. J Diabetes
Endocrinol Metab. 1973;37(5):826-828. Investig. Published online October 23, 2020. doi:10.1111/
8. Nauck M, Schmidt WE, Ebert R, et al. Insulinotropic proper- jdi.13446
ties of synthetic human gastric inhibitory polypeptide in man: 26. Hare KJ, Vilsbøll T, Holst JJ, Knop FK. Inappropriate glucagon

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


interactions with glucose, phenylalanine, and cholecystokinin-8. response after oral compared with isoglycemic intravenous
J Clin Endocrinol Metab. 1989;69(3):654-662. glucose administration in patients with type 1 diabetes. Am J
9. Mutt V, Jorpes JE, Magnusson S. Structure of porcine secretin. Physiol Endocrinol Metab. 2010;298(4):E832-E837.
The amino acid sequence. Eur J Biochem. 1970;15(3):513-519. 27. Nauck MA, Homberger E, Siegel EG, et al. Incretin effects of
10. Deckert T. Stimulation of insulin secretion by glucagon and se- increasing glucose loads in man calculated from venous in-
cretin. Acta Endocrinol (Copenh). 1968;57(4):578-584. sulin and C-peptide responses. J Clin Endocrinol Metab.
11. Fahrenkrug J, Schaffalitzky de Muckadell OB, Kühl C. Effect 1986;63(2):492-498.
of secretin on basal- and glucose-stimulated insulin secretion in 28. Bagger JI, Knop FK, Lund A, Vestergaard H, Holst JJ, Vilsbøll T.
man. Diabetologia. 1978;14(4):229-234. Impaired regulation of the incretin effect in patients with type 2
12. Perley MJ, Kipnis DM. Plasma insulin responses to oral and diabetes. J Clin Endocrinol Metab. 2011;96(3):737-745.
intravenous glucose: studies in normal and diabetic subjects. J 29. Madsbad S, Kehlet H, Hilsted J, Tronier B. Discrepancy between
Clin Invest. 1967;46(12):1954-1962. plasma C-peptide and insulin response to oral and intravenous
13. Nauck M, Stöckmann F, Ebert R, Creutzfeldt W. Reduced glucose. Diabetes. 1983;32(5):436-438.
incretin effect in type 2 (non-insulin-dependent) diabetes. 30. Horowitz M, Edelbroek MA, Wishart JM, Straathof JW.
Diabetologia. 1986;29(1):46-52. Relationship between oral glucose tolerance and gastric emptying
14. Ebert R, Unger H, Creutzfeldt W. Preservation of incretin ac- in normal healthy subjects. Diabetologia. 1993;36(9):857-862.
tivity after removal of gastric inhibitory polypeptide (GIP) 31. Holst JJ, Gribble F, Horowitz M, Rayner CK. Roles of the gut in
from rat gut extracts by immunoadsorption. Diabetologia. glucose homeostasis. Diabetes Care. 2016;39(6):884-892.
1983;24(6):449-454. 32. Jørgensen NB, Jacobsen SH, Dirksen C, et al. Acute and
15. Lauritsen KB, Moody AJ, Christensen KC, Lindkaer Jensen S. long-term effects of Roux-en-Y gastric bypass on glucose me-
Gastric inhibitory polypeptide (GIP) and insulin release tabolism in subjects with Type 2 diabetes and normal glucose
after small-bowel resection in man. Scand J Gastroenterol. tolerance. Am J Physiol Endocrinol Metab. 2012;303(1):E122
1980;15(7):833-840. -E131.
16. Holst JJ. The physiology of glucagon-like peptide 1. Physiol 33. Tolhurst G, Reimann F, Gribble FM. Nutritional regu-
Rev. 2007;87(4):1409-1439. lation of glucagon-like peptide-1 secretion. J Physiol.
17. Eissele R, Göke R, Willemer S, et al. Glucagon-like peptide-1 2009;587(1):27-32.
cells in the gastrointestinal tract and pancreas of rat, pig and 34. Kuhre RE, Frost CR, Svendsen B, Holst JJ. Molecular mechan-
man. Eur J Clin Invest. 1992;22(4):283-291. isms of glucose-stimulated GLP-1 secretion from perfused rat
18. Orskov C, Holst JJ, Knuhtsen S, Baldissera FG, Poulsen SS, small intestine. Diabetes. 2015;64(2):370-382.
Nielsen OV. Glucagon-like peptides GLP-1 and GLP-2, pre- 35. Færch K, Torekov SS, Vistisen D, et al. GLP-1 response to oral
dicted products of the glucagon gene, are secreted separately glucose is reduced in prediabetes, screen-detected type 2 dia-
from pig small intestine but not pancreas. Endocrinology. betes, and obesity and influenced by sex: the ADDITION-PRO
1986;119(4):1467-1475. study. Diabetes. 2015;64(7):2513-2525.
19. Mojsov S, Heinrich G, Wilson IB, Ravazzola M, Orci L, 36. Nauck MA, Vardarli I, Deacon CF, Holst JJ, Meier JJ. Secretion
Habener JF. Preproglucagon gene expression in pancreas and of glucagon-like peptide-1 (GLP-1) in type 2 diabetes: what is
intestine diversifies at the level of post-translational processing. up, what is down? Diabetologia. 2011;54(1):10-18.
J Biol Chem. 1986;261(25):11880-11889. 37. Holst JJ, Knop FK, Vilsbøll T, Krarup T, Madsbad S.
20. Holst JJ, Albrechtsen NJW, Gabe MBN, Rosenkilde MM. Loss of incretin effect is a specific, important, and
Oxyntomodulin: actions and role in diabetes. Peptides. early characteristic of type 2 diabetes. Diabetes Care.
2018;100:48-53. 2011;34(Suppl 2):S251-S257.
21. Holst JJ, Orskov C, Nielsen OV, Schwartz TW. Truncated 38. Knop FK, Vilsbøll T, Højberg PV, et al. Reduced incretin effect
glucagon-like peptide I, an insulin-releasing hormone from the in type 2 diabetes: cause or consequence of the diabetic state?
distal gut. FEBS Lett. 1987;211(2):169-174. Diabetes. 2007;56(8):1951-1959.
22. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon- 39. Eng J, Kleinman WA, Singh L, Singh G, Raufman JP. Isolation
like peptide-1 7-36: a physiological incretin in man. Lancet. and characterization of exendin-4, an exendin-3 analogue, from
1987;2(8571):1300-1304. Heloderma suspectum venom. Further evidence for an exendin
Endocrinology, 2021, Vol. 162, No. 7 9

receptor on dispersed acini from guinea pig pancreas. J Biol 55. Jørgensen NB, Dirksen C, Bojsen-Møller KN, et al. Exaggerated
Chem. 1992;267(11):7402-7405. glucagon-like peptide 1 response is important for improved
40. Raufman JP, Singh L, Eng J. Exendin-3, a novel peptide from β-cell function and glucose tolerance after Roux-en-Y gas-
Heloderma horridum venom, interacts with vasoactive intestinal tric bypass in patients with type 2 diabetes. Diabetes.
peptide receptors and a newly described receptor on dispersed 2013;62(9):3044-3052.
acini from guinea pig pancreas. Description of exendin-3(9-39) 56. Jepsen SL, Grunddal KV, Wewer Albrechtsen NJ, et al. Paracrine
amide, a specific exendin receptor antagonist. J Biol Chem. crosstalk between intestinal L- and D-cells controls secretion
1991;266(5):2897-2902. of glucagon-like peptide-1 in mice. Am J Physiol Endocrinol
41. Thorens B, Porret A, Bühler L, Deng SP, Morel P, Widmann C. Metab. 2019;317(6):E1081-E1093.
Cloning and functional expression of the human islet GLP-1 57. Gasbjerg LS, Helsted MM, Hartmann B, et al. Separate and
receptor. Demonstration that exendin-4 is an agonist and combined glucometabolic effects of endogenous glucose-
exendin-(9-39) an antagonist of the receptor. Diabetes. dependent insulinotropic polypeptide and glucagon-like peptide
1993;42(11):1678-1682. 1 in healthy individuals. Diabetes. 2019;68(5):906-917.

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


42. Chen YE, Drucker DJ. Tissue-specific expression of unique 58. Hansen LS, Sparre-Ulrich AH, Christensen M, et al.
mRNAs that encode proglucagon-derived peptides or exendin N-terminally and C-terminally truncated forms of glucose-
4 in the lizard. J Biol Chem. 1997;272(7):4108-4115. dependent insulinotropic polypeptide are high-affinity competi-
43. Schirra J, Sturm K, Leicht P, Arnold R, Göke B, Katschinski M. tive antagonists of the human GIP receptor. Br J Pharmacol.
Exendin(9-39)amide is an antagonist of glucagon- 2016;173(5):826-838.
like peptide-1(7-36)amide in humans. J Clin Invest. 59. Sparre-Ulrich AH, Gabe MN, Gasbjerg LS, et al. GIP(3-30)NH2
1998;101(7):1421-1430. is a potent competitive antagonist of the GIP receptor and ef-
44. Edwards CM, Todd JF, Mahmoudi M, et al. Glucagon-like pep- fectively inhibits GIP-mediated insulin, glucagon, and somato-
tide 1 has a physiological role in the control of postprandial statin release. Biochem Pharmacol. 2017;131:78-88.
glucose in humans: studies with the antagonist exendin 9-39. 60. Yanagimachi T, Fujita Y, Takeda Y, et al. Pancreatic glucose-
Diabetes. 1999;48(1):86-93. dependent insulinotropic polypeptide (GIP) (1-30) expression is
45. Elashoff M, Matveyenko AV, Gier B, Elashoff R, Butler PC. upregulated in diabetes and PEGylated GIP(1-30) can suppress
Pancreatitis, pancreatic, and thyroid cancer with glucagon-like the progression of low-dose-STZ-induced hyperglycaemia in
peptide-1-based therapies. Gastroenterology. 2011;141(1):150-156. mice. Diabetologia. 2016;59(3):533-541.
46. Ahmad SR, Swann J. Exenatide and rare adverse events. N Engl 61. Asmar M, Asmar A, Simonsen L, et al. The gluco- and
J Med. 2008;358(18):1970-1972. liporegulatory and vasodilatory effects of glucose-
47. Nauck MA. A critical analysis of the clinical use of incretin- dependent insulinotropic polypeptide (GIP) are abolished
based therapies: the benefits by far outweigh the potential risks. by an antagonist of the human GIP receptor. Diabetes.
Diabetes Care. 2013;36(7):2126-2132. 2017;66(9):2363-2371.
48. Salehi M, Vahl TP, D’Alessio DA. Regulation of islet hormone 62. Gasbjerg LS, Christensen MB, Hartmann B, et al. GIP(3-30)
release and gastric emptying by endogenous glucagon-like NH2 is an efficacious GIP receptor antagonist in humans: a ran-
peptide 1 after glucose ingestion. J Clin Endocrinol Metab. domised, double-blinded, placebo-controlled, crossover study.
2008;93(12):4909-4916. Diabetologia. 2018;61(2):413-423.
49. Adriaenssens AE, Svendsen B, Lam BY, et al. Transcriptomic 63. Nauck MA, Meier JJ. GIP and GLP-1: stepsiblings rather
profiling of pancreatic alpha, beta and delta cell populations than monozygotic twins within the incretin family. Diabetes.
identifies delta cells as a principal target for ghrelin in mouse 2019;68(5):897-900.
islets. Diabetologia. 2016;59(10):2156-2165. 64. Gasbjerg LS, Bergmann NC, Stensen S, et al. Evaluation of the
50. Xu SFS, Andersen DB, Izarzugaza JMG, Kuhre RE, Holst JJ. In incretin effect in humans using GIP and GLP-1 receptor antag-
the rat pancreas, somatostatin tonically inhibits glucagon secre- onists. Peptides. 2020;125:170183.
tion and is required for glucose-induced inhibition of glucagon 65. Højberg PV, Zander M, Vilsbøll T, et al. Near normalisation
secretion. Acta Physiol (Oxf). 2020;229(3):e13464. of blood glucose improves the potentiating effect of GLP-1 on
51. Kuhre RE, Christiansen CB, Ghiasi SM, et al. Neuromedin U glucose-induced insulin secretion in patients with type 2 dia-
does not act as a decretin in rats. Cell Metab. 2019;29(3):719- betes. Diabetologia. 2008;51(4):632-640.
726.e5. 66. Stensen S, Gasbjerg LS, Krogh LL, et al. Effects of endogenous
52. Svendsen B, Larsen O, Gabe MBN, et al. Insulin secre- GIP in patients with type 2 diabetes. Eur J Endocrinol.
tion depends on intra-islet glucagon signaling. Cell Rep. 2021;EJE-21-0135.R1.
2018;25(5):1127-1134.e2. 67. Aulinger BA, Bedorf A, Kutscherauer G, et al. Defining the role
53. Serre V, Dolci W, Schaerer E, et al. Exendin-(9-39) is an in- of GLP-1 in the enteroinsulinar axis in type 2 diabetes using
verse agonist of the murine glucagon-like peptide-1 re- DPP-4 inhibition and GLP-1 receptor blockade. Diabetes.
ceptor: implications for basal intracellular cyclic adenosine 2014;63(3):1079-1092.
3′,5′-monophosphate levels and β-cell glucose competence. 68. Vilsbøll T, Knop FK, Krarup T, et al. The pathophysiology of
Endocrinology. 1998;139(11):4448-4454. diabetes involves a defective amplification of the late-phase in-
54. Svendsen B, Holst JJ. Paracrine regulation of somatostatin se- sulin response to glucose by glucose-dependent insulinotropic
cretion by insulin and glucagon in mouse pancreatic islets. polypeptide-regardless of etiology and phenotype. J Clin
Diabetologia. 2021;64(1):142-151. Endocrinol Metab. 2003;88(10):4897-4903.
10  Endocrinology, 2021, Vol. 162, No. 7

69. Kjems LL, Holst JJ, Vølund A, Madsbad S. The influence of 74. Højberg PV,Vilsbøll T, Rabøl R, et al. Four weeks of near-normalisation
GLP-1 on glucose-stimulated insulin secretion: effects on beta- of blood glucose improves the insulin response to glucagon-like pep-
cell sensitivity in type 2 and nondiabetic subjects. Diabetes. tide-1 and glucose-dependent insulinotropic polypeptide in patients
2003;52(2):380-386. with type 2 diabetes. Diabetologia. 2009;52(2):199-207.
70. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W. 75. Frias JP, Nauck MA, Van J, et al. Efficacy and safety of
Normalization of fasting hyperglycaemia by exogenous glucagon- LY3298176, a novel dual GIP and GLP-1 receptor agonist, in
like peptide 1 (7-36 amide) in type 2 (non-insulin-dependent) dia- patients with type 2 diabetes: a randomised, placebo-controlled
betic patients. Diabetologia. 1993;36(8):741-744. and active comparator-controlled phase 2 trial. Lancet.
71. Jones AG, McDonald TJ, Shields BM, et al; PRIBA Study 2018;392(10160):2180-2193.
Group. Markers of β-cell failure predict poor glycemic response 76. Killion EA, Wang J, Yie J, et al. Anti-obesity effects
to GLP-1 receptor agonist therapy in type 2 diabetes. Diabetes of GIPR antagonists alone and in combination with
Care. 2016;39(2):250-257. GLP-1R agonists in preclinical models. Sci Transl Med
72. Toft-Nielsen MB, Madsbad S, Holst JJ. Determinants of the ef- 2018;10(472):eaat3392.

Downloaded from https://academic.oup.com/endo/article/162/7/bqab065/6199910 by guest on 23 January 2023


fectiveness of glucagon-like peptide-1 in type 2 diabetes. J Clin 77. Willard FS, Douros JD, Gabe MB, et al. Tirzepatide is an
Endocrinol Metab. 2001;86(8):3853-3860. imbalanced and biased dual GIP and GLP-1 receptor agonist.
73. Billings LK, Doshi A, Gouet D, et al. Efficacy and safety of JCI Insight 2020;5(17):e140532.
IDegLira versus basal-bolus insulin therapy in patients with 78. Min T, Bain SC. The role of tirzepatide, dual GIP and
type 2 diabetes uncontrolled on metformin and basal in- GLP-1 receptor agonist, in the management of type 2
sulin: the DUAL VII randomized clinical trial. Diabetes Care. diabetes: the SURPASS clinical trials. Diabetes Ther.
2018;41(5):1009-1016. 2021;12(1):143-157.

You might also like