You are on page 1of 20

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/359722934

Inhibition of Insulin Degrading Enzyme to Control Diabetes Mellitus and its


Applications on some Other Chronic Disease: a Critical Review

Article  in  Pharmaceutical Research · April 2022


DOI: 10.1007/s11095-022-03237-7

CITATIONS READS

0 70

5 authors, including:

Md. Shofiul Azam Md Wahiduzzaman


Dhaka University of Engineering & Technology Shanghai Institute of Nutrition and Health Chinese Academy of Sciences
22 PUBLICATIONS   108 CITATIONS    2 PUBLICATIONS   0 CITATIONS   

SEE PROFILE SEE PROFILE

Md. Reyad-ul- Ferdous Md. Nahidul Islam


Shandong University Bangabandhu Sheikh Mujibur Rahman Agricultural University
83 PUBLICATIONS   300 CITATIONS    66 PUBLICATIONS   769 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

formulation and development of levetiracetam film coated tablets View project

Anticancer Medicinal Plants View project

All content following this page was uploaded by Md Wahiduzzaman on 06 April 2022.

The user has requested enhancement of the downloaded file.


Pharmaceutical Research
https://doi.org/10.1007/s11095-022-03237-7

EXPERT REVIEW

Inhibition of Insulin Degrading Enzyme to Control Diabetes Mellitus


and its Applications on some Other Chronic Disease: a Critical Review
Md. Shofiul Azam1   · Md. Wahiduzzaman2 · Md. Reyad‑ul‑Ferdous3 · Md. Nahidul Islam4 · Mukta Roy5

Received: 15 January 2022 / Accepted: 14 March 2022


© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2022

Abstract
Purpose  This review aims to provide a precise perceptive of the insulin-degrading enzyme (IDE) and its relationship to type
2 diabetes (T2D), Alzheimer’s disease (AD), obesity, and cardiovascular diseases. The purpose of the current study was to
provide clear idea of treating prevalent diseases such as T2D, and AD by molecular pharmacological therapeutics rather
than conventional medicinal therapy.
Methods  To achieve the aims, molecular docking was performed using several softwares such as LIGPLOT+, Python, and
Protein-Ligand Interaction Profiler with corresponding tools.
Results  The IDE is a large zinc-metalloprotease that breakdown numerous pathophysiologically important extracellular
substrates, comprising amyloid β-protein (Aβ) and insulin. Recent studies demonstrated that dysregulation of IDE leads
to develop AD and T2D. Specifically, IDE regulates circulating insulin in a variety of organs via a degradation-dependent
clearance mechanism. IDE is unique because it was subjected to allosteric activation and mediated via an oligomer structure.
Conclusion  In this review, we summarised the factors that modulate insulin reformation by IDE and interaction of IDE and
some recent reports on IDE inhibitors against AD and T2D. We also highlighted the latest signs of progress of the function
of IDE and challenges in advancing IDE- targetted therapies against T2D and AD.

KEY WORDS  amyloid-peptide · IDE · insulin · T2D and Alzheimer’s disease

Introduction (1). IDE quantified to the 110 kDa zinc protease of the M16
family, which is involved in the degradation and release of
Insulin protease and insulinase is an insulin-degrading highly preserved insulin (2, 3), amyloid-β (Aβ) peptides
enzyme (IDE), a zinc metalloprotease that attached and (4), Insulin-like growth factor II (IGF-II) (5), glucagon (6),
changed several types of bioactive peptides by correspond- amylin (7) as well as somatostatin (8). It plays an active
ing their sequences and structures, inhibiting peptide aggre- role in reducing glucagon, β-amyloid, and chemokine ligand
gation formation to the numerous subcellular compartments 3, among other peptide substrates, through a high insulin

2
* Md. Shofiul Azam Bio‑Med Big Data Center, CAS Key Laboratory
shofiul@duet.ac.bd of Computational Biology, CAS‑MPG Partner Institute
for Computational Biology, Shanghai Institute of Nutrition
Md. Wahiduzzaman
and Health, University of Chinese Academy of Sciences,
wahid@picb.ac.cn
Chinese Academy of Sciences, Shanghai 200031, China
Md. Reyad‑ul‑Ferdous 3
Department of Endocrinology and Metabolism, Shandong
rockyreyad@sdu.edu.cn
Provincial Hospital affiliated to Shandong University,
Md. Nahidul Islam Shandong University, Jinan 250021, Shandong, China
nahidul.islam@bsmrau.edu.bd 4
Department of Agro‑Processing, Bangabandhu Sheikh
Mukta Roy Mujibur Rahman Agricultural University, Gazipur 1706,
muktaroy-fet@sust.edu Bangladesh
5
1 Department of Food Engineering and Tea Technology,
Department of Chemical and Food Engineering, Dhaka
Shahjalal University of Science and Technology,
University of Engineering & Technology, Gazipur 1707,
Sylhet 3114, Bangladesh
Bangladesh

13
Vol.:(0123456789)
Pharmaceutical Research

sensitivity. The IDE was present in the cellular membranes, mechanisms of insulin-degrading enzymes and their roles in
cytoplasm, and several cell organelles (peroxisomes, mito- pathogenesis and discusses the potential therapeutic options
chondria, and endosomes) and collected from the extracel- based on IDE inhibition in human diseases.
lular region (9–11). Also, it is settled in Escherichia coli
from mammals and is expressed in both insulin-sensitive/
non-sensitive cells, intimating a multifunctional protein (9). Role of Insulin‑Degrading Enzyme (IDE)
IDE was discovered by Arthur Mirsky in 1949. Mirsky
argued that IDE’s inhibitors would be a perfect anti-diabetic Working Mechanism of Insulin‑Degrading Enzyme
medication, allowing them to limit insulin breakdown. Mir- (IDE)
sky also discovered that when the liver brought out the
capacity of an IDE inhibitor and administered it to rabbits, In the enzymatic model that refers to 130/159 atoms, the
they improved insulin action. Mirsky and others explored to IDE performs its catalytic action on two peptides of distinct
produce efficacious IDE inhibitors as potential medications lengths using zinc-binding proteases, mimicking a part of
(12, 13). the B chain of insulin, using concentration functional theo-
IDE inhibitor depreciated the hypoglycemia action of retical methods and hybrid exchange at the gas stage and
insulin, indicating potential therapeutic against IDE (14). interactions in protein environments effective is B3LYP (26).
Despite these attempts, only a few chemicals that selectively According to the authors ‘crystal structures, IDE matches
block IDE are now accessible, except IDE substrates such as a clamshell, besides two bowl-shaped sections joined using
insulin (15). The active site of the IDE reveals in unparal- an amenable hinge (Fig. 1). The protease can transit between
leled aspect the crystal structure imparted by Noinaj, et al. open and closed states with this arrangement. The elongated
(16), which holds the key to carrying through a significant hydrogen bonding on both parts of IDE provides a “latch”
and all-embracing IDE inhibitor. Insulin activity demon- that tends to remain the peptide closed (Fig. 1A), according
strated to be potentiated by IDE inhibitors both in cultured to Shen, et al. (28). Researchers were able to boost the effi-
cells and in vivo. Highly selective IDE inhibitors showed cacy of peptides in cleaving a test substrate by up to 40-fold
substantial anti-diabetic benefits attributed to decreased by addressing the mutations to the enzyme that disrupts the
insulin catabolism (17). hydrogen-bond make fast (Fig. 1B). The breakdown of insu-
The implications of apolipoprotein E (APOE) risk alleles lin and amyloid protein was efficient (13).
on cerebral, lipid, and cholesterol metabolism leading to So, what’s the mechanism behind the mutant IDE’s pro-
CVD-mediated insulin resistance (IR) are highlighted, as nounced enzyme activation? Consider a basic double stair
well as the consequences of cardiovascular risk factors model of the enzyme response to understand that. At first,
leading to IR and amyloid formation. IR, genetic, and car- an enzyme-substrate complex developed by combining
diovascular risk factors all play a role in amyloid aggrega- enzymes and substrates in the reverse process. Then, the
tion, which can lead to the late onset of AD and DM (18). substrate is catalytically cleaved while the reaction products
Interleukin-6 (IL-6) is a proinflammatory cytokine that con- are simultaneously released. Mutations that boost the pro-
trols differentiation, migration, proliferation, and cell death tease’s open state, approve binding substrate for increasing
and hence plays a key role in the development of insulin the reaction’s productivity by speeding up the creation of the
resistance and the pathogenesis of type 2 diabetes mellitus enzyme-substrate complex (29).
(T2DM) (19). Impairment of IDE function is associated with
the pathophysiology of both type-2-diabetes (T2D) and Alz- Source and Nomenclature of IDE
heimer’s disease (AD), according to the atlas database from
human and animal modeling studies as well as molecular Although well known as IDE insulin, it is a highly preserved
epigenetics (20–22). The IDE is a prototypical member of M16A ­Zn2+ metalloprotein developed in the tissues of bacte-
the zinc-metal prototype of a distinct superfamily that has ria, fungi, plants, animals, and humans brain, liver, and mus-
various characteristics that differentiate it from the ordinary cles that were first selected by its capacity to rapidly break
metallic prototypes, having a zinc-binding motif (HxxEH) down insulin (30). IDE is unique becasue it has a strong
(inverted) (23) and an unusual tertiary structure (24). affinity for substrates with a wide range of sequences and
The subcellular localization of IDE is another distinctive structures. The 200 remnants of the N-terminal also con-
feature: in the cytosol, most of the massive IDE is found, tain some inverted zinc-metalloprotease core motifs (e.g.,
with minor aggregates found in the mitochondria, peroxi- HXXEH). This is why IDE is classified as a member of
somes, and endosomes. The tiny amount of IDE ranges the “inverzincins” family. It’s a thiol zinc-metalloendopepti-
between 3% to 10% transported to the extracellular space, dase with a molecular weight of 110 kDa that’s found in
where it communicates with recognized IDE substrates like the cytosol, peroxisomes, endosomes, and cell surface (31).
insulin and Aβ (25). This study focuses on the molecular Insulin, amylin, and β-amyloid protein are among the tiny

13
Pharmaceutical Research

Fig. 1  Insulin-degrading enzyme (IDE) activation pathway. (a) Insulin-degrading enzyme (IDE) is an enzyme that breaks down compounds
linked to Alzheimer’s disease and diabetes. The crystal form of IDE showed a, ‘latch’ mechanism (Red) that holds the enzyme in a closed state,
delaying the entry of the substrate or exit of the cleavage products. (b) Mutations (Blue) that disrupt the latch promote the open conformation of
the enzyme. All of the mutations are more willing to receive the substrates and discharge consequences the typically appearing the IDE, which is
more active (27).

proteins that IDE catalyzes the breakdown of insulin (32). complexes, and the surface of cerebrovascular endothelial
IDE homologs play the most vital role in the selection of bud cells, its roles may be reversed (36).
sites and mating in budding yeast (33). The IDE features a
one-of-a-kind regulation mechanism. It has an inferior for- The Substrate Recognition and Catalytic Mechanism
mulation and sphere organization with yeast mitochondria of IDE
refining proteinase (17).
All the structural solutions of IDE in complex with the insu-
Prospect of IDE Degradation on Insulin lin B-chain, glucagon, and amylin shed light on how IDE
interacts with its substrate and how it cleaves. When bound
The ability of IDE to damage the A and B chains of the to IDE, all three substrates have a total of 70 residues and
hormone insulin, primarily at the locations Leu13-Tyr14 fit entirely within the crypt, transforming from a -helix to a
and Tyr14-Gln15 (A chain) and Ser9-His10, His10-Leu11, -strand shape (28). The substrate can engage non-covalently
Glu13-Ala14, Tyr15-Leu16, and Phe25-Tyr26 (B chain), (B with two IDE-N areas: the catalytic site and the postulated
chain was first discovered). This result revealed that IDE exosite. The N-terminus of the substratum is attached to the
and pitrilysin, a bacterial protease, have a high degree of exosite of the enzyme by engaging with a sheet of IDE-N
similarity and a likely similar proteolytic process. In terms once it has been trapped (28). The exosite is a highly con-
of primary sequence, the M16A subfamily enzymes are very served area of IDE that may have a regulatory role in sub-
similar. About 30% of identity exists between human IDE strate selectivity and catalysis (28, 37). This connection acts
and pitrilysin. This resemblance can be found during the in the way that a molecular chain, allowing the C-terminal
whole of the length of selected proteins; however, it is com- end of the substrate to be properly positioned at the catalytic
monly more prominent in the N-terminal portion (34). region where cleavage occurs (38).
Some researchers looked at mice that didn’t have any The presence of disulfide bonds is a distinguishing feature
activity in the insulysin gene. They discovered that the of some IDE substrates (Fig. 2). Insulin, for example, has
insulysin enzyme destroys amyloid peptides as well as insu- three disulfide bonds. The cleavage of insulin by IDE, sur-
lin. Moreover, even a little reduction in insulysin activity prisingly, does not necessitate the decrease and disintegra-
raised amyloid peptide levels in the brain (35). The involve- tion of these disulfide bonds. Several other IDE substrates
ment of IDE in Alzheimer’s disease is now well understood; (e.g., amylin, insulin growth factor I and II) have disulfide
nevertheless, the biological roles of IDE and many other linkages, which could affect how they interact with the cata-
M16A enzymes are still poorly understood. Because IDE is lytic chamber of the enzyme. However, disulfide linkages
found in the cytosol, peroxisomes, endosomes, proteasome are not present in all of IDE’s substrates (e.g., glucagon,

13
Pharmaceutical Research

Fig. 2  Human native and


recombinant IDEs are depicted
schematically. The proteom-
ics sequence inferred from the
human IDE cDNA sequence is
referred to as amino acid posi-
tions (GenBank Accession No.
M21188). Where amino acid
sequences that are found in both
natural and recombinant IDEs
are represented with hatched
boxes. The sequence between
the first putative initiation
methionine (Met 1) and IDE’s
likely the start codon which is
denoted by the black box (Met
42). The tag sequences of the
recombinant IDEs are repre-
sented by the shadow color and
open boxes.

amyloid-β). It’s unclear how the presence of disulfide bonds reach the active site in a closed structure because it is
affects substrate interaction in the crypt, and therefore more stored in chambers formed by the two concave domains.
research is needed (39). The closed-to-open transfer of catalytic conversion cav-
ity facilitates entry into the middle layer. A significant
IDE Crystal Structure boost (approximately 40-fold) in the competent catalytic
of the enzyme is disclosed if mutations inhibit the closed
The active site of zinc-binding is kept within a proteolytic conformation (37).
cavity produced by defined N- terminal and C-terminal This finding showed that altering IDE’s structural prefer-
units linked with a loop including 28 amino acid residues, ence to the open state could be a potential Alzheimer’s treat-
according to studies aimed at determining the crystal struc- ment. This should speed up the breakdown of Aβ, preventing
ture of IDE. The catalytic chamber is large enough to bind aggregation and neuron death (40).
and destroy insulin-like amyloid (A) layers, which contain
amino acid residues 39–43 and respectively (39). The Function of IDE as a Protease of Insulin
In these circumstances, the active site anchors the N-ter-
minus of peptides with an exosite that is about 30 degrees The ability of the IDE to impair insulin into multiple frag-
out of the way from the catalytic core. This allows the sub- ments in vitro, giving the principle and trivial outcomes,
strates to be properly located, allowing cleavage at the cata- was its first distinguishing feature. The first cleavage
lytic site. However, because smaller peptides cannot bind the events take place in the center of the insulin A and B
exosite and the catalytic core simultaneously, the anchoring chains, with no necessary particular amino acids, implying
and degrading mechanism for substrates less than around that IDE recognizes substrates in terms of the sequence of
12 amino acids are unknown. In any case, a researcher has amino acid on the tertiary structure rather than primary
found that the percentages of catalysis of IDE are higher (41). As a competitive inhibitor, the poor hydrolysis sub-
for small substrates (> 2000 ­s−1) than for lengthy substrates stratum is proinsulin which is sustained at slow rates, but
likely insulin (about 0.56 min −1) (39). IDE has a high sympathy for insulin (Km 0.1 M) (42).
Insulin-like growth factor I (IGF-I) and IGF-II are both
Configuration of IDE IDE substrates, with IGF-II degrading faster than IGF-I.
Despite this, they were operating as IDE inhibitors (42).
There are two possible configurations for the IDE. The The leader peptide of rat prethiolase B (P27 peptide),
first is an open conformation, which lets substrates and found in peroxisomes, is another powerful inhibitor of
products enter and depart. The other is more difficult to IDE’s insulin-degrading activity (43).

13
Pharmaceutical Research

Molecular and Biochemical Characteristics of IDE insulin counteraction. In Diet-Induced Obese (DIO) mice,
pioglitazone increased IDE activity that declared a mod-
A gene on the human chromosome 10 q23–q25 and mouse ern algorithm of thiazolidinedione act in insulin resist-
chromosome 19 give rise to IDE as a unique polypeptide ance treatment. The data indicated that the high-fat diet
with a molecular weight of approximately 110 kDa. In the enhanced the IDE activity in the liver of DIO mice and
Goto–Kakizaki rat model, IDE coding alterations have been IDE’s movement control by substrate-specific inhibitors
linked to the development of T2D (15). Fakhrai-Rad, et al. (15) (46, 47). Pioglitazone induced IDE’s movement, implying
discovered two missense variants in IDE (H18R and A890V) a novel medicinal effect in the treatment of T2D.
that reduce the ability of transfected cells to digest insulin by
31%. They found that the two mutations had a synergistic
effect on insulin breakdown that is perplexing conferred, where Induction of IDE Protein by Glucagon and Forskolin
H18R is existent in the intentional mitochondrial region of
IDE generated via substitute translation initiation (10). Farris, The cells are processed with forskolin, a small molecule
et al. (44) discovered that recombinant IDE with the A980V protein kinase A (PKA) activator, which showed a similar
mutation alone had a lower level of catalytic ability for both model of IDE protein completion that shows the gluca-
insulin and Aβ degradation, implying that this mutation is par- gon promoted IDE protein via the cAMP/PKA signaling
ticularly important for the proteolytic function. Intriguingly, pathway. Forskolin’s PKA pathway had the analogous gain
there was no payoff on insulin breakdown in cell lysates from as glucagon. As a result, the cAMP pathway is probably
IDE-transfected COS cells, implying the outcome is contingent obstructed in glucagon-induced IDE upregulation (47).
on ligand-receptor hormone internalization (43).
Inhibition of IDE Protein by TNF‑α
IDE Is Heat Shock Protein
TNF-α raised IDE protein moderately at one hour, then dropped
HSR is a genetic response to a variety of environmental and dramatically after that in the 8-h treatment. In hepatocytes,
physiological stresses that cause the encoding of genes to TNF-α decreased IDE mRNA. TNF-α seems to have a consid-
incorporate molecular chaperone, proteases, and other pro- erable inhibitory effect on IDE expression in hepatocytes (48).
teins (mostly from the heat shock gene superfamily) that TNF-α increases insulin resistance in adipocytes and peripheral
incorporate and incorrectly combine proteins needed to pro- tissues via affecting insulin signaling via serine phosphoryla-
tect against cellular damage. The Hsp100, Hsp90, Hsp70, tion, leading to the development of T2DM (49, 50).
Hsp60, Hsp40 (J-domain proteins), with short heat shock
protein (sHSP) lineages are arranged in terms of molecular IDE Inhibitors
structure with a magnitude of category in the heat shock gene
superfamily. The maximum number of small molecules act IDE inhibitors were first discovered in 1949 and allowed
namely molecular chaperones, causing structural changes to treat T2DM (51). Mirsky, and his colleagues, identi-
which are necessary for protein synthesis, folding, transloca- fied IDE endogenous inhibitors from rat tissue extracts
tion, assembly, and destruction. Most of the HSPs are stabi- (52). Partly purified insulin-degrading protease function
lizing detachable proteins and resell deformed proteins under they entitled insulinase, the precise identification of these
stressful conditions. If proteins are irrevocably switched, inhibitors remained unknown. Nevertheless, Mirsky and col-
molecular chaperones assist in their transfer to the cell’s pro- leagues employed functional properties of IDE inhibitors to
teolytic machinery, namely the ubiquitin-proteasome path- evaluate the hypothesis that IDE antagonists might be an
way. We show that IDE behaves like a heat shock protein and alternative to treat diabetes inhibit the endogenous insulin
that up-regulated amounts of the enzyme can be seen in CNS breakdown rather than injected exogenous insulin (53). A
malignancies in vivo. According to the findings, IDE performs recent study has been found a physiologically active mac-
a vital role in neuroblastoma cell proliferation via interacting rocycle that carefully inhibits the IDE in diabetes mellitus
with the ubiquitin-proteasome system (30). patients, potentially providing fresh therapy alternatives.
The inhibitor significantly enhanced glucose tolerance and
slowed stomach emptying in both lean and obese animals.
The inhibitor modulated circulation levels of glucagon and
Factors that Affect IDE Activity amylin, two hormones that are regulated by glucose and
blood sugar levels, in the view of collection to increase insu-
Zinc stimulates IDE enzyme activity, while copper, alu- lin levels in mice. The anti-diabetic action of IDE inhibitors
minium, and nitric oxide hinder it (45). In mice, knock- is mediated by several hormones, according to the original
ing down the IDE gene causes hyperinsulinemia and article (54).

13
Pharmaceutical Research

It has been reported that non-proteinaceous insulinase activity, substratum opposition of unlabeled to labeled insu-
inhibitors exhibited hypoglycemic effects both in rabbit and lin, enzyme inhibitor outline, and visualization by a par-
rodent models (53). Taken together, several antidiabetic drug ticular antibody were all used to characterize the enzyme.
moieties have been developed since the 1950s (55–57). Insulin breakdown was inhibited dose-dependently by 1 to
Several studies have been conducted to develop nontarget 5 mM ATP (Trichloroacetic acid determined by precipitation
specific IDE used like thiol-alkylating compounds, zinc-chela- and insulin antibody binding). 3 mM adenosine 5-diphos-
tors, cyclic peptide bacitracin, and cyclic peptide bacitracin to phate, adenosine 5-monophosphate, guanosine 5-triphos-
access the functional effect of IDE in insulin catabolism before phate, pyrophosphate, −methyleneadenosine 5-triphosphate,
the development of selective IDE inhibitors. Non-specific IDE adenosine 5-O-(3thiotriphosphate), and dibutiryl cyclic
inhibitors suppress insulin degradation in rat L6 myoblasts, adenosine 5-monophosphate inhibited 74%, 4%, 38%, 46%,
mouse BC3H1 muscle cells, and HepG2 cells (58–60). 65%, 36%, respectively and because the plot of 1/v (insu-
Although robust and rationally selective, Ii1 owned lin degradation) versus ATP concentration non-linear and
the drawback of synthesizing. Leissring and colleagues the Hill coefficient was greater than one, a kinetic study of
developed a series of truncated variants, including a 455- ATP inhibition suggested an allosteric impact (1.51 and
Da allyl ester comprising only the 2-Nap and Arg resi- 2.44) (Fig. 3). The allosteric inhibition binding constant
dues to achieve the goal (61). Surprisingly, these modi- was Ki T = 1.5107  M, indicating a decrease in enzyme
fications exhibited unusual substrate selectivity, turning affinity caused by ATP. We concluded that ATP inhibits the
in potency by up to 300-fold for various substrates, not enzyme’s insulin breakdown activity (63).
existing actives it directed compounds (61). Abdul-Hay,
et al. (61) reported Peptide Hydroxamate (MW > 740) is Role of Metal Ions in IDE
target specific IDE inhibitor, which is difficult for chemical
synthesis. This study revealed that, in at least a few cases, Biometals like copper, aluminium, and zinc play an impor-
the inhibition of IDE might depend on tertiary interac- tant part including Alzheimer’s disease and diabetes melli-
tions amongst the enzyme, substrate, and inhibitor, lend- tus. Zinc stimulates IDE enzyme activity, while copper, alu-
ing assistance to the feasibility of uncovering substrate- minium, and nitric oxide hinder it. In mice, knocking down
selective drug moieties (Table I). the IDE gene causes hyperinsulinemia and insulin resistance
(35, 64, 65). The findings of several experimental techniques
Effect of Hepatic IDE on Insulin reveal that copper (I) inhibits IDE irreversibly, but that it can
still process its substrates when linked to copper (II) (66).
Insulin dictation of IDE operation is intriguing since fat Metal ions have the potential to influence IDE proteolytic
causes hyperinsulinemia. The relationship is built on the dif- activity in at least three different ways: Metal ions can trig-
ferent systems. To find the answer to this query, a research ger conformational changes on IDE substrates, which can
investigation looked at IDE’s movement in 1c1c7 cells after result in proteolytic resistance forms. In this regard, zinc,
pushing the insulin. For 8 h, the cells were given 200 nM but not copper, has been shown to cause the development
insulin. In both protein and mRNA, IDE activity was meas- of A aggregates that are resistant to IDE proteolytic activity
ured. In the insulin-treated cells, no significant change in IDE (67, 68). External influences, such as high metal concen-
expression was found, implying that insulin does not affect the trations that encourage Aβ aggregate formation, may con-
straightway of IDE’s expression in hepatocytes (48). Under tribute to Aβ buildup by lowering the peptide’s sensitivity
a liver cell model, researchers reported insulin regulates IDE to proteolytic degradation; ii. Direct metal ion binding to
activity and this regulation is lost in high glucose circum- IDE may affect the enzyme’s function. Metal ions have long
stances. This disruption in IDE protein expression or splicing been known to help enzymes stay stable and active (69). The
cannot be explained by corresponding changes. Furthermore, binding of metal cofactors in various oxidation states, such
it has been found that insulin engagement is associated with as calcium, magnesium, manganese, copper, zinc, and iron,
the rise in IDE mRNA levels as well as a trend toward higher has been demonstrated to boost the thermal stabilities of
IDE protein levels in high glucose circumstances (62). proteins (70) and to have a significant impact on their struc-
ture and activity (71). Modulation of IDE’s proteolytic activ-
ATP Inhibits Insulin‑Degrading Enzyme Activity ity could thus occur via a variety of mechanisms, including
exogenous ligand coordination, substitution, or removal
The ability of ATP to suppress the IDE enzyme’s degrading of the catalytic metal (72). Furthermore, metals may bind
activity in vitro was investigated. Several chromatographic to residues in the active site, preventing substrate interac-
methods apply to purify the enzyme from rat skeletal mus- tion, or to residues outside the active site, compromising
cle. In a polyacrylamide gel, the final purification stage the enzyme’s structural integrity (71); iii. In vivo, several
revealed two bands of 110 and 60 kDa. Insulin breakdown metals may influence IDE expression and localization (66).

13
Pharmaceutical Research

Table I  Structure and common name of insulin degrading enzyme, IDE (Protein/receptor) Potential inhibitor candidates.
Compoun Lipinski’s rule of five
Molecul
d Name / Molecular structure and Referenc
ar Pocket Binding(3D)
ID Interaction with IDE(2D) Properties Value es
formula
Number

Molecular weight
(<500 639.1
Da)

LogP (<5) 2

C42 H86
li1 (103)
O3

H-Bond donor (5) 3

H-bond acceptor (<10) 37

Molecular weight
(<500 757.9
Da)

LogP (<5) 3.5

C41H55N
6bK (104)
7O7

H-Bond donor (5) 7

H-bond acceptor (<10) 8

13
Pharmaceutical Research

Table I  (continued)

Molecular weight
(>500 733.9
Da)

LogP (>5) 7.8

C44H55N (105,106
NTE-1
5O5 )

H-Bond donor (5) 3

H-bond acceptor (<10) 7

Molecular weight
(<500 447.5
Da)

LogP (<5) 2.5

BDM4476 C24H22F
(14)
8 N5O3

H-Bond donor (5) 3

H-bond acceptor (<10) 6

13
Pharmaceutical Research

Table I  (continued)

Molecular weight
(<500 378
Da)

LogP (<5) 0

15349090
B35 (107)
4

H-Bond donor (5) 0

H-bond acceptor (<10) 7

Molecular weight
(<500 479.5
Da)

LogP (<5) 1.6

C21H22F
ML345 (108)
N3O5S2

H-Bond donor (5) 0

H-bond acceptor (<10) 9

13
Pharmaceutical Research

Table I  (continued)

Molecular weight
(<500 735.9
Da)

LogP (<5) 8.6

C44H54F
NTE-2 (109)
N5O4

H-Bond donor (5) 3

H-bond acceptor (<10) 7

Molecular weight
(<500 319.9
Da)

LogP (<5) 4.6

Quinoline C18H26C
(110)
2 lN3

H-Bond donor (5) 1

H-bond acceptor (<10) 3

13
Pharmaceutical Research

Table I  (continued)

Molecular weight
(<500 375.4
Da)

LogP (<5) 5.6

C23H15F
Brequinar (111)
2NO2

H-Bond donor (5) 1

H-bond acceptor (<10) 5

Molecular weight
(<500 324.4
Da)

LogP (<5) 2.9

C20H24N
Quinine (112)
2O2

H-Bond donor (5) 1

H-bond acceptor (<10) 4

­ IGPLOT+, Python, and Protein-Ligand Interaction Profiler with corresponding


The study of molecular docking required software names are L
tools.

Fig. 3  Effect of ATP on IDE


on T2D.

13
Pharmaceutical Research

Role of Metformin in IDE processed more efficiently. Memory and learning prob-
lems are linked to somatostatin inanition in the cortex and
Several literature findings suggested that in addition to hippocampus of Alzheimer’s patients as well as the devel-
Aβ synthesis and conveyance, several signalling path- opment of Alzheimer’s disease appears to be linked to a
ways could entangle in metformin’s impact. In both APP significant decrease of somatostatin in the mouse CA1
(amyloid precursor protein, Mo/HuAPP695swe) and hippocampus region. Somatostatin appears to represent a
PS1(a mutant human presenilin 1) transgenic mice, the role of the principal activity of neprilysin via influencing
anti-diabetic medication metformin was found to success- its expression and synaptic location (78).
fully alleviate AD symptoms. In the brains of both APP
and PS1 mice, Glucophage arouses AMPK and enhances
IDE. The metformin could activate IDE to ameliorate IDE Application in some Therapeutic
Aβ-induced pathology, mainly AD (73). Diseases
In older persons, sporadic cerebral amyloid angiopa-
thy (CAA), marked by cerebrovascular Aβ deposits, pro- Schematic diagram for application of IDE in different
duces brain haemorrhages and dementia. Metformin has chronic diseases shown in Fig. 5.
been used to treat T2DM patients, and animal and clinical
research have shown that it can help people with Alz- Inhibition Compared to Activation of IDE
heimer’s disease (Fig. 4). Metformin reduced the sever-
ity of CAA by increasing Aβ-cleaving IDE expression. Recent discoveries raise doubts about the wisdom of shut-
The clinical application of metformin in CAA treatment ting down the IDE. The discovery that IDE physically
may lead to a novel therapeutic strategy, particularly in destroys the amyloid protein which saves up pathologi-
patients with T2D (74–76). cally in Alzheimer’s disease is one of the most signifi-
cant. Rather than inhibiting IDE, it would be preferable
to activate it, a method that has already been shown to
Resveratrol Sustains IDE be useful in mice approaches of the particular disorder.
Furthermore, studies from many animal patterns put sus-
When compared to the control, resveratrol causes a sig- picion on the idea of permanently blocking IDE to cure
nificant increase in Aβ42 fragmentation, indicating the diabetics (12).
polyphenol promotes IDE contingent decay of Aβ42 and Despite intensive research into IDE’s role as an insulin
its parts. We observed that implying resveratrol and IDE- protease, there is little evidence that it plays a significant
based treatment and prevention methods for sporadic AD impact on hepatic insulin clearance in vivo. The effects
may be promising (77). of IDE inhibitors on insulin clearance in vivo, such as the
NTE-1 inhibitor, have not been studied or have shown
no effect. The following are the fundamental proof that
Somatostatin Modulates Insulin‑Degrading‑Enzyme implicates IDE in hepatic endosomal insulin proteolysis:
Metabolism first, its bluff relation for insulin, implying the selec-
tive enzyme for the hormone; second, the discovery that
Somatostatin (SST) is a substrate and allosteric regulator cleavage insulin feedback achieved from isolated hepatic
of IDE mobility, allowing a synthetic Aβ-peptide to be endosomes are similar to those produced by purified

Fig. 4  Effect of metformin on
IDE to treat AD.

13
Pharmaceutical Research

IDE; and third, the detection of small pools of IDE in insulin clearance function. Insulin clearance is mediated by
endosomes (81). IDE, which is a significant rate-limiting enzyme. In gene
knockout mice, inactivation of IDE activity causes hyperin-
Mechanism of IDE in Liver Regulation sulinemia. Insulin and numerous intermediate-size peptides
are degraded by IDE. In diabetic patients, both insulin clear-
In an 8-h investigation, pioglitazone (5 mM) elevated ance and IDE activity are diminished, while the cause-effect
IDE protein and mRNA in a time-reliant way in the link in humans is unknown (81).
mouse hepatoma cell line (Hepa 1c1c7 cells), reveal- Because of IDE’s parts in insulin clearance, IDE inhibi-
ing the formulation of IDE standing order in the liver. tors are utilized to control insulin levels in T2D patients
The presence of a free fatty acid (palmitate 300 mM) (83). IDE created certain specific insulin fragments that
increased IDE’s small molecule but decreased mRNA. have been found to have peculiar biological effects that are
IDE protein was reduced by glucagon and TNF-α. Insu- not evident for the entire hormone under the experimental
lin showed no operation in the same circumstance. We settings used. The optimum IDE inhibitor for treating T2D
can conclude that pioglitazone and FFA directly boosted should prioritize insulin and amylin clearance while leav-
IDE activity in hepatocytes, but Glucagon and TNF-α ing other IDE substrates, such as glucagon, unaffected (17).
reduced it (43, 48).

Effect of IDE on Diabetics Treatment and Insulin Advance Prospect of IDE against Alzheimer’s


Clearance Disease (AD)

The liver is an important part of insulin clearance, as it is a. Gene therapy. Insulin signalling and IDE levels are
responsible for eliminating 70% of the insulin produced on reduced in mice fed a high-fat diet, indicating a physi-
the islet (82). Hyperinsulinemia is caused by a decrease in ological relationship in an AD animal model. Increased

Fig. 5  Schematic diagram for


application of IDE in differ-
ent chronic diseases. T2DM,
insulin resistance, obesity,
and neurodegeneration are all
symptoms of T2DM. In both
T2DM and AD, metabolic
excess, chronic inflammation,
and oxidative stress cause cel-
lular dysfunction. Because brain
IR can arise without diabetes,
it’s possible that Alzheimer’s
disease develops in the early
phases of insulin resistance.
Chronic inflammation and
oxidative stress are thought to
be two main mechanisms that
link diabetes and Alzheimer’s
disease (79, 80).

13
Pharmaceutical Research

Fig. 6  IDE inhibitor prevents disruption of different proteins.

IDE gene dosage has been shown to prevent and treat They uncovered a carboxyterminal RB amino acid sequence
the development of AD pathology in a transgenic that resembled the catalytic core of IDE ten years later. This
mouse model (84, 85). As gene therapy biotechnology structural similarity indicates that insulin degradation is a
advances, this strategy could become a viable treatment common tumor-suppressive mechanism in both RB and IDE.
option for AD. These findings suggest that methods to Following that, a first immunohistochemistry examination of
improve insulin signalling could be employed as a treat- the differential expression of human IDE in normal tissues,
ment for Alzheimer’s disease (20, 86). Furthermore, IDE primary tumors, and their associated lymph node metastases
increasing insulin signalling, and increasing IDE would backed up the initial hypothesis that IDE was an anticancer
avoid the issue of lower insulin neurotrophic effects molecule. According to this study, IDE contains ankyrin
induced by elevated IDE (23, 87). repeat-like amino acid sequences through which it might bind
b. Induction of enzymes. Hersh’s team discovered a new and so inhibit the pro-inflammatory factor NF-kappaB and
peptide that stimulates IDE activity. Other research cyclin-dependent kinases (CDKs). As seen above, IDE also
groups are looking for comparable compounds as well. has two RXL cyclin-binding domains, which could contribute
This method is less invasive and maybe the best strategy to its supposed suppression of CDKs. These new findings
to prevent Alzheimer’s disease. imply that IDE may decrease inflammation and oncogenesis
c. T2D treatment. At present urgent demand is required through a variety of pathways that ensure RB function.
against insulin resistance for the subset of T2D patients This finding suggests a precise method for regulating IDE
with hyperinsulinemia rather than taking exogenous activity and/or that there are yet-to-be-identified IDE natural
insulin. Reduced insulin level leads to an increased level inhibitors that could be misregulated, at least in this tumor.
of IDE to initiate degradation, lowering the likelihood Nestin, a type VI intermediate filament protein expressed
of Alzheimer’s disease (88). by numerous cell types throughout development and at high
d. Increasing the PPAR/AMPK signalling pathway to levels by tumor cells such as neuroblastoma and glioma
treat T2D and AD. The activators of the PPAR/AMPK cells, has been discovered to prevent monomeric ubiquitin
signalling pathway dramatically raised the expression breakdown by IDE (30, 89, 90).
level of IDE in mice with T2D and AD. They reduced
the buildup of A40 and A42, as well as the impairments IDE Is Involved in Ubiquitin Clearance and Varicella
in spatial learning and recognition. To some extent, Zoster Virus Infection
PPAR and AMPK activators could be used to treat peo-
ple with T2D and Alzheimer’s disease (54). Chickenpox and frequently reactivates in adulthood is
caused by the varicella-zoster virus (VZV) virus, a neu-
rotropic human herpes virus and frequently reactivates in
IDE for Tumor Suppressor and Anti‑Inflammatory adulthood, resulting in zoster or, less commonly, cerebral
Protein vasculitis. There have been studies linking IDE to the
etiology of various classification of physiopathological
In 1994, the researcher hypothesized that IDE inhibits malig- processes, including diseases, in the last decade, includ-
nant transformation by degrading insulin, preventing this main ing associating the protein with a wide range of cellular
growth-stimulatory hormone from attaching to the retinoblas- processes, including VZV infection, steroid receptor sign-
toma tumor suppressor protein (RB) and so inactivating it. aling, and insulin-dependent proteasome modulation. It’s

13
Pharmaceutical Research

also been suggested that IDE plays a role in rat muscle Conclusion
cell differentiation and development, implying that it has
various cellular growth and tissue homeostasis functions. The IDE is a multifunctional protein that leads initiate sev-
Furthermore, recent discoveries suggest that (i) IDE is eral pathological conditions in humans. The therapeutic
involved in the process of antigenic peptides presented strategies targeting IDE could be effective in treating sev-
by class I MHC via a proteasome-independent pathway, eral diseases. IDE breaks down insulin and amylin, both
and (ii) ubiquitin can be destroyed by IDE “in vitro,” even of which are linked to T2D, as well as amyloid-β(Aβ), a
though this last conclusion is somewhat contradictory. peptide involved in Alzheimer’s disease pathology. Hyper-
Finally, preliminary research illustrated that in neural insulinemia and T2D develop Alzheimer’s disease in the
cells attacked with viruses capable of shutting down cel- senior citizen due to enzyme activity, including substrate
lular proteins, the content of IDE was either preserved affinities. We believe that a lack of IDE causes an increase in
or even increased, demonstrating HSP-like action, with Aβ and thus AD pathology in the brain. Although the study
an HSP-like behavior, IDE content was conserved or was conducted in liver hepatocytes, the findings may apply
even increased. All of these findings point to IDE being to IDE control in other tissues including kidneys, brain, and
engaged in ubiquitin-proteasome pathways, and hence in adipose tissues. Whereas, emerging IDE inhibitors against
the response related to heat shock (HSR), compelling us T2D, and AD are in the earlier drug discovery stage, reveal-
to look into this possibility (30). ing IDE structure and function will illustrate new insight
into drug discovery to treat IDE-related diseases.

The IDE Plays Multifunctional Protein ACKNOWLEDGMENTS AND DISCLOSURES  We thank Dr. Dang Fabin
from Beth Israel Deaconess Medical Center (BIDMC), Harvard for
the critical reading of this manuscript.
Many physiologically relevant amyloidogenic peptides,
including insulin, IGF-II, TGF-, glucagon, amylin, atrial Authors’ Contribution  Md. Shofiul Azam conceived the idea and
natriuretic peptide (ANP), calcitonin, β-endorphin, A b, wrote the original manuscript, writing review and editing. Md. Wahi-
amyloid Bri (ABri), amyloid Dan (ADan), and the C-ter- duzzaman analyzed the molecular docking, writing review, and edit-
minal domain of A b precursor protein (APP-AICD), ing, Md. Reyad-ul-Ferdous, Md. Nahidul Islam, and Mukta Roy
writing review and editing. All authors have read and agreed to the
somatostatin, are reduced mono shown in Fig. 6 (8, 46). As current version of the manuscript.
a result, poisonous aggregates are prevented from forming,
leading to a variety of clinical disorders (91–93). Data Availability  Protein and Ligand ID accession numbers were
However, data suggests that IDE functions not just collected from CRSB (Protein Data Bank) and PubChem databases
through proteolysis but also interactions with other respectively. Molecular docking study required software names are
LIGLOT+, Python, and Protein-Ligand Interaction Profiler with cor-
intracellular proteins (94, 95), containing amyloido- responding tools.
genic peptides with chaperone-like function (96, 97) and
α-synuclein (α-syn), an amyloidogenic peptide involved in Declarations 
Parkinson’s disease (98, 99). By binding to androgen and
glucocorticoid receptors and boosting their DNA binding Ethics Approval and Consent to Participate  This study does not relate
capacity, IDE may have a role in the cross-talk between to any human and animal trial.
insulin/insulin-like growth factors and steroid hormone
Consent for Publication  The author declares no conflict of interest.
signalling pathways (100).
Competing Interests  Not applicable.
Coronary Heart Disease

In the Chinese Han population, IDE gene polymorphisms


are strongly linked to the likelihood of developing coronary References
heart disease (CHD). Caravaggio, et al. (101) investigated
Ldlr−/− mice and found that the reduced IDE levels in the 1. Malito E, Hulse RE, Tang WJ. Amyloid beta-degrading crypti-
mice increased the levels of Aβ and AGEs, thus inducing dases: insulin degrading enzyme, presequence peptidase, and
neprilysin. Cell Mol Life Sci. 2008;65(16):2574–85.
inflammation and atherosclerosis. This clinical study found
2. Hersh LB. The insulysin (insulin degrading enzyme) enigma.
that IDE gene polymorphisms were linked to the develop- Cell Mol Life Sci. 2006;63(21):2432–4.
ment of atherosclerotic lesions, including CHD (102). 3. Fernández-Gamba A, Leal MC, Morelli L, Castaño EM.
Insulin-degrading enzyme: structure-function relationship

13
Pharmaceutical Research

and its possible roles in health and disease. Curr Pharm Des. 22. Carrasquillo MM, Belbin O, Zou F, Allen M, Ertekin-Taner
2009;15(31):3644–55. N, Ansari M, Wilcox SL, Kashino MR, Ma L, Younkin LH,
4. Kurochkin IV, Goto S. Alzheimer's beta-amyloid peptide specifi- Younkin SG, Younkin CS, Dincman TA, Howard ME, Howell
cally interacts with and is degraded by insulin degrading enzyme. CC, Stanton CM, Watson CM, Crump M, Vitart V, et al. Con-
FEBS Lett. 1994;345(1):33–7. cordant Association of Insulin Degrading Enzyme Gene (IDE)
5. Guo Q, Manolopoulou M, Bian Y, Schilling AB, Tang WJ. variants with IDE mRNA, Aß, and Alzheimer's disease. PLoS
Molecular basis for the recognition and cleavages of IGF-II, One. 2010;5(1):e8764.
TGF-alpha, and amylin by human insulin-degrading enzyme. J 23. Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ubeda
Mol Biol. 2010;395(2):430–43. OJ, Frautschy SA, Cole GM. Insulin-degrading enzyme as
6. Fo A, Cameron PH, Cm M, Kouach M, Briand G. Endosomal a downstream target of insulin receptor signaling cascade:
proteolysis of glucagon at neutral pH generates the bioactive implications for Alzheimer's disease intervention. J Neurosci.
degradation product Miniglucagon-(19–29). Endocrinology. 2004;24(49):11120–6.
2003;144(12):5353–64. 24. Huang W, Ramsey KM, Marcheva B, Bass J. Circadian rhythms,
7. Bennett RG, Hamel FG, Duckworth WC. An insulin-degrad- sleep, and metabolism. J Clin Invest. 2011;121(6):2133–41.
ing enzyme inhibitor decreases amylin degradation, increases 25. Vitaterna MH, Takahashi JS, Turek FW. Overview of circadian
amylin-induced cytotoxicity, and increases amyloid formation rhythms. Alcohol Res Health. 2001;25(2):85–93.
in insulinoma cell cultures. Diabetes. 2003;52(9):2315–20. 26. Amata O, Marino T, Russo N, Toscano M. Human insulin-
8. Ciaccio C, Tundo GR, Grasso G, Spoto G, Marasco D, Ruvo M, degrading enzyme working mechanism. J Am Chem Soc.
Gioia M, Rizzarelli E, Coletta M. Somatostatin: a novel substrate 2009;131(41):14804–11.
and a modulator of insulin-degrading enzyme activity. J Mol 27. Leissring MA, Selkoe DJ. Enzyme target to latch on to. Nature.
Biol. 2009;385(5):1556–67. 2006;443(7113):761–2.
9. Duckworth WC, Bennett RG, Hamel FG. Insulin degradation: 28. Shen Y, Joachimiak A, Rosner MR, Tang WJ. Structures of
progress and potential. Endocr Rev. 1998;19(5):608–24. human insulin-degrading enzyme reveal a new substrate recog-
10. Leissring MA, Farris W, Wu X, Christodoulou DC, Haigis MC, nition mechanism. Nature. 2006;443(7113):870–4.
Guarente L, Selkoe DJ. Alternative translation initiation gener- 29. Elsässer B, Goettig P. Mechanisms of proteolytic enzymes
ates a novel isoform of insulin-degrading enzyme targeted to and their inhibition in QM/MM studies. Int J Mol Sci.
mitochondria. Biochem J. 2004;383(Pt. 3):439–46. 2021;22(6):3232.
11. Farris W, Leissring MA, Hemming ML, Chang AY, Selkoe DJ. 30. Tundo GR, Sbardella D, Ciaccio C, Bianculli A, Orlandi A,
Alternative splicing of human insulin-degrading enzyme yields Desimio MG, Arcuri G, Coletta M, Marini S. Insulin-degrading
a novel isoform with a decreased ability to degrade insulin and enzyme (IDE): a novel heat shock-like protein. J Biol Chem.
amyloid β-protein. Biochemistry. 2005;44(17):6513–25. 2013;288(4):2281–9.
12. Leissring MA, Farris W, Chang AY, Walsh DM, Wu X, Sun X, 31. Song ES, Rodgers DW, Hersh LB. Insulin-degrading enzyme is
Frosch MP, Selkoe DJ. Enhanced proteolysis of beta-amyloid not secreted from cultured cells. Sci Rep. 2018;8(1):2335.
in APP transgenic mice prevents plaque formation, secondary 32. Son SM, Kang S, Choi H, Mook-Jung I. Statins induce insulin-
pathology, and premature death. Neuron. 2003;40(6):1087–93. degrading enzyme secretion from astrocytes via an autophagy-
13. Leissring MA, Selkoe DJ. Structural biology: enzyme target to based unconventional secretory pathway. Mol Neurodegener.
latch on to. Nature. 2006;443:761+. 2015;10:56.
14. Deprez-Poulain R, Hennuyer N, Bosc D, Liang WG, Enée E, 33. Galagovsky D, Katz MJ, Acevedo JM, Sorianello E, Glavic A,
Marechal X, Charton J, Totobenazara J, Berte G, Jahklal J, Wappner P. The Drosophila insulin-degrading enzyme restricts
Verdelet T, Dumont J, Dassonneville S, Woitrain E, Gauriot growth by modulating the PI3K pathway in a cell-autonomous
M, Paquet C, Duplan I, Hermant P, Cantrelle F-X, et al. Cata- manner. Mol Biol Cell. 2014;25(6):916–24.
lytic site inhibition of insulin-degrading enzyme by a small 34. Sharma SK, Chorell E, Steneberg P, Vernersson-Lindahl E,
molecule induces glucose intolerance in mice. Nat Commun. Edlund H, Wittung-Stafshede P. Insulin-degrading enzyme pre-
2015;6(1):8250. vents α-synuclein fibril formation in a nonproteolytical manner.
15. Fakhrai-Rad H, Nikoshkov A, Kamel A, Fernström M, Zierath Sci Rep. 2015;5:12531.
JR, Norgren S, Luthman H, Galli J. Insulin-degrading enzyme 35. Bellia F, Pietropaolo A, Grasso G. Formation of insulin frag-
identified as a candidate diabetes susceptibility gene in GK rats. ments by insulin-degrading enzyme: the role of zinc(II) and cys-
Hum Mol Genet. 2000;9(14):2149–58. tine bridges. J Mass Spectrometry. 2013;48(2):135–40.
16. Noinaj N, Song E-S, Bhasin S, Alper B, Schmidt W, Hersh L, 36. Carpenter JE, Jackson W, de Souza GA, Haarr L, Grose C. Insu-
Rodgers D. Anion activation site of insulin-degrading enzyme. lin-degrading enzyme binds to the nonglycosylated precursor
J Biol Chem. 2011;287:48–57. of varicella-zoster virus gE protein found in the endoplasmic
17. Tang W-J. Targeting insulin-degrading enzyme to treat type 2 reticulum. J Virol. 2010;84(2):847–55.
diabetes mellitus. Trends Endocrinol Metab. 2016;27(1):24–34. 37. Im H, Manolopoulou M, Malito E, Shen Y, Zhao J, Neant-Fery
18. Jabeen K, Rehman K, Akash MSH. Genetic mutations of M, Sun CY, Meredith SC, Sisodia SS, Leissring MA, Tang WJ.
APOEε4 carriers in cardiovascular patients lead to the devel- Structure of substrate-free human insulin-degrading enzyme
opment of insulin resistance and risk of Alzheimer's disease. J (IDE) and biophysical analysis of ATP-induced conformational
Biochem Mol Toxicol. 2022;36(2):e22953. switch of IDE. J Biol Chem. 2007;282(35):25453–63.
19. Rehman K, Akash MSH, Liaqat A, Kamal S, Qadir MI, Rasul A. 38. Grasso G, Bush AI, D’Agata R, Rizzarelli E, Spoto G. Enzyme
Role of Interleukin-6 in development of insulin resistance and solid-state support assays: a surface plasmon resonance and mass
type 2. Diabetes Mellitus. 2017;27(3):229–36. spectrometry coupled study of immobilized insulin degrading
20. Kurochkin IV, Guarnera E, Berezovsky IN. Insulin-degrading enzyme. Eur Biophys J. 2008;38(4):407.
enzyme in the fight against Alzheimers disease. Trends Pharma- 39. Hulse RE, Ralat LA, Wei-Jen T. Structure, function, and regula-
col Sci. 2018;39(1):49–58. tion of insulin-degrading enzyme. Vitam Horm. 2009;80:635–48.
21. Qiu WQ, Folstein MF. Insulin, insulin-degrading enzyme and 40. Çakir B, Dağliyan O, Dağyildiz E, Bariş İ, Kavakli IH, Kizilel
amyloid-beta peptide in Alzheimer's disease: review and hypoth- S, Türkay M. Structure based discovery of small molecules to
esis. Neurobiol Aging. 2006;27(2):190–8.

13
Pharmaceutical Research

regulate the activity of human insulin degrading enzyme. PLoS 57. Marigo S, Panelli G. Insulinase and its inhibition by hypoglyce-
One. 2012;7(2):e31787. mic sulfonamides; data on insulin sensitivity during tolbutamide
41. Manolopoulou M, Guo Q, Malito E, Schilling AB, Tang W-J. therapy. Arch Sci Med. 1958;105(6):587–609.
Molecular basis of catalytic chamber-assisted unfolding and 58. Gehm BD, Rosner MR. Regulation of insulin, epidermal growth
cleavage of human insulin by human insulin-degrading enzyme. factor, and transforming growth factor-alpha levels by growth fac-
J Biol Chem. 2009;284(21):14177–88. tor-degrading enzymes. Endocrinology. 1991;128(3):1603–10.
42. Misbin RI, Almira EC, Duckworth WC, Mehl TD. Inhibition of 59. Kayalar C, Wong WT. Metalloendoprotease inhibitors which
insulin degradation by insulin-like growth factors. Endocrinol- block the differentiation of L6 myoblasts inhibit insulin degrada-
ogy. 1983;113(4):1525–7. tion by the endogenous insulin-degrading enzyme. J Biol Chem.
43. González-Casimiro CM, Merino B, Casanueva-Álvarez E, 1989;264(15):8928–34.
Postigo-Casado T, Cámara-Torres P, Fernández-Díaz CM, 60. Kayalar C, Wong WT, Hendrickson L. Differentiation of BC3H1
Leissring MA, Cózar-Castellano I, Perdomo G. Modulation of and primary skeletal muscle cells and the activity of their endog-
insulin sensitivity by insulin-degrading enzyme. Biomedicines. enous insulin-degrading enzyme are inhibited by the same metal-
2021;9(1):86. loendoprotease inhibitors. J Cell Biochem. 1990;44(3):137–51.
44. Farris W, Mansourian S, Leissring MA, Eckman EA, Bertram 61. Abdul-Hay SO, Lane AL, Caulfield TR, Claussin C, Bertrand
L, Eckman CB, Tanzi RE, Selkoe DJ. Partial loss-of-function J, Masson A, Choudhry S, Fauq AH, Maharvi GM, Leissring
mutations in insulin-degrading enzyme that induce diabetes also MA. Optimization of peptide Hydroxamate inhibitors of insulin-
impair degradation of amyloid &#x3b2;-protein. Am J Pathol. degrading enzyme reveals marked substrate-selectivity. J Med
2004;164(4):1425–34. Chem. 2013;56(6):2246–55.
45. Leissring MA, Malito E, Hedouin S, Reinstatler L, Sahara T, 62. Pivovarova O, Gögebakan Ö, Pfeiffer AFH, Rudovich N. Glucose
Abdul-Hay SO, Choudhry S, Maharvi GM, Fauq AH, Huzarska inhibits the insulin-induced activation of the insulin-degrading
M, May PS, Choi S, Logan TP, Turk BE, Cantley LC, Manolo- enzyme in HepG2 cells. Diabetologia. 2009;52(8):1656–64.
poulou M, Tang W-J, Stein RL, Cuny GD, Selkoe DJ. Designed 63. Camberos MC, Perez AA, Udrisar DP, Wanderley MI, Cresto JC.
inhibitors of insulin-degrading enzyme regulate the catabolism ATP inhibits insulin-degrading enzyme activity. Exp Biol Med.
and activity of insulin. PLoS One. 2010;5(5):e10504. 2001;226(4):334–41.
46. Maianti JP, McFedries A, Foda ZH, Kleiner RE, Du XQ, 64. Perlman RK, Rosner MR. Identification of zinc ligands of the
Leissring MA, Tang WJ, Charron MJ, Seeliger MA, Saghat- insulin-degrading enzyme. J Biol Chem. 1994;269(52):33140–5.
elian A, Liu DR. Anti-diabetic activity of insulin-degrading 65. Bellia F, Grasso G. The role of copper(II) and zinc(II) in the
enzyme inhibitors mediated by multiple hormones. Nature. degradation of human and murine IAPP by insulin-degrading
2014;511(7507):94–8. enzyme. J Mass Spectrom. 2014;49(4):274–9.
47. Maianti JP, Tan GA, Vetere A, Welsh AJ, Wagner BK, Seel- 66. Grasso G, Salomone F, Tundo GR, Pappalardo G, Ciaccio C,
iger MA, Liu DR. Substrate-selective inhibitors that reprogram Spoto G, Pietropaolo A, Coletta M, Rizzarelli E. Metal ions
the activity of insulin-degrading enzyme. Nat Chem Biol. affect insulin-degrading enzyme activity. J Inorg Biochem.
2019;15(6):565–74. 2012;117:351–8.
48. Wei X, Ke B, Zhao Z, Ye X, Gao Z, Ye J. Regulation of insu- 67. Bellia F, Lanza V, Ahmed IMM, Garcia-Vinuales S, Veiss
lin degrading enzyme activity by obesity-associated factors and E, Arizzi M, Calcagno D, Milardi D, Grasso G. Site directed
pioglitazone in liver of diet-induced obese mice. PLoS One. mutagenesis of insulin-degrading enzyme allows singling out
2014;9(4):e95399. the molecular basis of peptidase versus E1-like activity: the role
49. Akash MSH, Rehman K, Liaqat A. Tumor necrosis factor-alpha: of metal ions. Metallomics. 2019;11(2):278–81.
role in development of insulin resistance and pathogenesis of 68. Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk
type 2 diabetes mellitus. J Cell Biochem. 2018;119(1):105–10. M, MacGregor L, Kiers L, Cherny R, Li Q-X, Tammer A, Car-
50. Faheem A, Rehman K, Jabeen K, Akash MSH. Nicotine-medi- rington D, Mavros C, Volitakis I, Xilinas M, Ames D, Davis S,
ated upregulation of microRNA-141 expression determines Beyreuther K, Tanzi RE, Masters CL. Metal-protein attenuation
adipokine-intervened insulin resistance. Environ Toxicol Phar- with Iodochlorhydroxyquin (Clioquinol) targeting Aβ amyloid
macol. 2020;80:103506. deposition and toxicity in Alzheimer disease: a pilot phase 2
51. Mirsky IA, Broh-Kahn RH. The inactivation of insulin by tissue clinical trial. Arch Neurol. 2003;60(12):1685–91.
extracts; the distribution and properties of insulin inactivating 69. Hamel FG, Bennett RG, Duckworth WC. Regulation of multi-
extracts. Arch Biochem. 1949;20(1):1–9. catalytic enzyme activity by insulin and the insulin-degrading
52. Mirsky IA, Simkin B, Broh-Kahn RH. The inactivation of insulin enzyme. Endocrinology. 1998;139(10):4061–6.
by tissue extracts. VI. The existence, distribution and properties 70. Kim N, Lee HJ. Redox-active metal ions and amyloid-
of an insulinase inhibitor. Arch Biochem. 1950;28(3):415–23. degrading enzymes in Alzheimer’s disease. Int J Mol Sci.
53. Mirsky IA, Perisutti G. Effect of insulinase-inhibitor on 2021;22(14):7697.
hypoglycemic action of insulin. Science (New York, NY). 71. Hadden JM, Déclais A-C, Phillips SEV, Lilley DMJ. Metal ions
1955;122(3169):559–60. bound at the active site of the junction-resolving enzyme T7
54. Li H, Wu J, Zhu L, Sha L, Yang S, Wei J, Ji L, Tang X, Mao K, endonuclease I. EMBO J. 2002;21(13):3505–15.
Cao L, Wei N, Xie W, Yang Z. Insulin degrading enzyme con- 72. Soulière MF, Perreault JP, Bisaillon M. Characterization of the
tributes to the pathology in a mixed model of Type 2 diabetes vaccinia virus D10 decapping enzyme provides evidence for a
and Alzheimer's disease: possible mechanisms of IDE in T2D two-metal-ion mechanism. Biochem J. 2009;420(1):27–35.
and AD. Biosci Rep. 2018;38(1):BSR20170862. 73. Lu X-Y, Huang S, Chen Q-B, Zhang D, Li W, Ao R, Leung FC-Y,
55. Mirsky IA, Perisutti G, Gitelson S. The role of insulinase in the Zhang Z, Huang J, Tang Y, Zhang S-J. Metformin ameliorates Aβ
hypoglycemic response to sulfonylureas. Ann N Y Acad Sci. pathology by insulin-degrading enzyme in a transgenic mouse
1957;71(1):103–11. model of Alzheimer’s disease. Oxidative Med Cell Longev.
56. Leites SM, Smirnov NP. The importance of the insulin inactivat- 2020;2020:2315106.
ing properties of the liver (insulinase) in the mechanism of action 74. Inoue Y, Masuda T, Misumi Y, Ando Y, Ueda M. Metformin
of antidiabetic sulfonamide preparations. Bull Exp Biol Med. attenuates vascular pathology by increasing expression of
1959;47(6):711–4. insulin-degrading enzyme in a mixed model of cerebral

13
Pharmaceutical Research

amyloid angiopathy and type 2 diabetes mellitus. Neurosci Lett. 93. Chiti F, Dobson CM. Protein misfolding, functional amyloid, and
2021;762:136136. human disease. Annu Rev Biochem. 2006;75:333–66.
75. Khalil MR, Ebeid A, Fayed H, Abd-Elhady S. Metformin: new 94. Bennett RG, Fawcett J, Kruer M, Duckworth W, Hamel FG.
insights into Alzheimer disease protection. Asian. J Biochem. Insulin inhibition of the proteasome is dependent on degrada-
2019;15(1):21–7. tion of insulin by insulin-degrading enzyme. J Endocrinol.
76. Rotermund C, Machetanz G, Fitzgerald JC. The therapeutic 2003;177(3):399–405.
potential of metformin in neurodegenerative diseases. Front 95. Epting CL, King FW, Pedersen A, Zaman J, Ritner C, Bernstein
Endocrinol. 2018;9:400. HS. Stem cell antigen-1 localizes to lipid microdomains and
77. Krasinski CA, Ivancic VA, Zheng Q, Spratt DE, Lazo ND. Res- associates with insulin degrading enzyme in skeletal myoblasts.
veratrol sustains insulin-degrading enzyme activity toward Aβ42. J Cell Physiol. 2008;217(1):250–60.
ACS Omega. 2018;3(10):13275–82. 96. Llovera RE, De Tullio M, Alonso LG, Leissring MA, Kaufman
78. Tundo G, Ciaccio C, Sbardella D, Boraso M, Viviani B, Coletta SB, Roher AE, de Prat GG, Morelli L, Castaño EM. The catalytic
M, Marini S. Somatostatin modulates insulin-degrading-enzyme domain of insulin-degrading enzyme forms a denaturant-resistant
metabolism: implications for the regulation of microglia activity complex with amyloid β peptide: implications for Alzheimer dis-
in AD. PLoS One. 2012;7(4):e34376. ease pathogenesis. J Biol Chem. 2008;283(25):17039–48.
79. Rehman K, Akash MSH. Mechanisms of inflammatory 97. de Tullio MB, Castelletto V, Hamley IW, Martino Adami PV,
responses and development of insulin resistance: how are they Morelli L, Castaño EM. Proteolytically inactive insulin-degrad-
interlinked? J Biomed Sci. 2016;23(1):87. ing enzyme inhibits amyloid formation yielding non-neurotoxic
80. Tegg M. Plasma insulin-degrading enzyme: Characterisation Aβ peptide aggregates. PLoS One. 2013;8(4):e59113.
and evaluation as a potential biomarker for Alzheimer's dis- 98. De Kimpe L, Scheper W. From alpha to omega with Aβ: targeting
ease.Masters Thesis. In: School of Medical Sciences: Edith the multiple molecular appearances of the pathogenic peptide in
Cowan University. Joondalup, Australia; 2014. Alzheimer's disease. Curr Med Chem. 2010;17(3):198–212.
81. Leissring MA, González-Casimiro CM, Merino B, Suire CN, 99. Steneberg P, Bernardo L, Edfalk S, Lundberg L, Backlund
Perdomo G. Targeting insulin-degrading enzyme in insulin F, Östenson C-G, Edlund H. The type 2 diabetes–associated
clearance. Int J Mol Sci. 2021;22(5):2235. gene ide is required for insulin secretion and suppression of
82. Costes S, Butler PC. Insulin-degrading enzyme inhibition, a novel α-synuclein levels in β-cells. Diabetes. 2013;62(6):2004–14.
therapy for type 2 diabetes? Cell Metab. 2014;20(2):201–3. 100. Tundo GR, Sbardella D, Ciaccio C, Grasso G, Gioia M,
83. Nash Y, Ganoth A, Borenstein-Auerbach N, Levy-Barazany H, Coletta A, Polticelli F, Di Pierro D, Milardi D, Van Endert P,
Goldsmith G, Kopelevich A, Pozyuchenko K, Sakhneny L, Laz- Marini S, Coletta M. Multiple functions of insulin-degrading
don E, Blanga-Kanfi S, Alhadeff R, Benromano T, Landsman enzyme: a metabolic crosslight? Crit Rev Biochem Mol Biol.
L, Tsfadia Y, Frenkel D. From virus to diabetes therapy: char- 2017;52(5):554–82.
acterization of a specific insulin-degrading enzyme inhibitor for 101. Caravaggio JW, Hasu M, MacLaren R, Thabet M, Raizman
diabetes treatment. FASEB J. 2021;35(5):e21374. JE, Veinot JP, Marcel YL, Milne RW, Whitman SC. Insulin-
84. Del Campo M, Stargardt A, Veerhuis R, Reits E, Teunissen CE. degrading enzyme deficiency in bone marrow cells increases ath-
Accumulation of BRI2-BRICHOS ectodomain correlates with a erosclerosis in LDL receptor-deficient mice. Cardiovasc Pathol.
decreased clearance of Aβ by insulin degrading enzyme (IDE) in 2013;22(6):458–64.
Alzheimer's disease. Neurosci Lett. 2015;589:47–51. 102. Cai P, Zhong W, Jia M, Yu C-q, Peng Y, Wang Y, Wang H-y,
85. Pivovarova-Ramich O, Höhn A, Grune T, Pfeiffer A, Rudovich Zeng C, Bai Y, Wang X. The gene polymorphisms of insulin
N. Insulin-degrading enzyme: new therapeutic target for diabetes degrading enzyme ( IDE ) are associated with the risk of coro-
and Alzheimer’s disease? Ann Med. 2016;48:1–11. nary heart disease in Chinese Han population. In.; 2016.
86. Zou F, Carrasquillo MM, Pankratz VS, Belbin O, Morgan K, 103. Rocha NKR, Themoteo R, Brentani H, Forlenza OV, De Paula
Allen M, Wilcox SL, Ma L, Walker LP, Kouri N, Burgess JD, VJR. Neuronal-glial interaction in a triple-transgenic mouse
Younkin LH, Younkin SG, Younkin CS, Bisceglio GD, Crook model of Alzheimer's disease: gene ontology and Lithium path-
JE, Dickson DW, Petersen RC, Graff-Radford N, et al. Gene ways. Front Neurosci. 2020;14:579984.
expression levels as endophenotypes in genome-wide associa- 104. Hahn F, Schmalen A, Setz C, Friedrich M, Schlößer S, Kölle J,
tion studies of Alzheimer disease. Neurology. 2010;74(6):480–6. Spranger R, Rauch P, Fraedrich K, Reif T, Karius-Fischer J, Bal-
87. Ferdous MR, Azam MS. Cardiac disease: current approaches asubramanyam A, Henklein P, Fossen T, Schubert U. Proteolysis
to gene therapy. In: Bulletin of Medical and Clinical Research; of mature HIV-1 p6 gag protein by the insulin-degrading enzyme
2020. p. 62–75. (IDE) regulates virus replication in an Env-dependent manner.
88. Qiu C, Gelaye B, Denis M, Tadesse MG, Luque Fernandez MA, PLoS One. 2017;12(4):e0174254.
Enquobahrie DA, Ananth CV, Sanchez SE, Williams MA. Circa- 105. Nunez LR, Jesch SA, Gaspar ML, Almaguer C, Villa-Garcia M,
dian clock-related genetic risk scores and risk of placental abrup- Ruiz-Noriega M, Patton-Vogt J, Henry SA. Cell wall integrity
tion. Placenta. 2015;36(12):1480–6. MAPK pathway is essential for lipid homeostasis. J Biol Chem.
89. Ralat LA, Kalas V, Zheng Z, Goldman RD, Sosnick TR, Tang 2008;283(49):34204–17.
WJ. Ubiquitin is a novel substrate for human insulin-degrading 106. Durham TB, Toth JL, Klimkowski VJ, Cao JX, Siesky AM,
enzyme. J Mol Biol. 2011;406(3):454–66. Alexander-Chacko J, Wu GY, Dixon JT, McGee JE, Wang Y,
90. Joly N, Engl C, Jovanovic G, Huvet M, Toni T, Sheng X, Stumpf Guo SY, Cavitt RN, Schindler J, Thibodeaux SJ, Calvert NA,
MP, Buck M. Managing membrane stress: the phage shock pro- Coghlan MJ, Sindelar DK, Christe M, Kiselyov VV, et al. Dual
tein (Psp) response, from molecular mechanisms to physiology. exosite-binding inhibitors of insulin-degrading enzyme challenge
FEMS Microbiol Rev. 2010;34(5):797–827. its role as the primary mediator of insulin clearance in vivo. J
91. Mukherjee A, Morales-Scheihing D, Butler PC, Soto C. Type Biol Chem. 2015;290(33):20044–59.
2 diabetes as a protein misfolding disease. Trends Mol Med. 107. Wang T, Yang N, Liang C, Xu H, An Y, Xiao S, Zheng M, Liu
2015;21(7):439–49. L, Wang G, Nie L. Detecting protein-protein interaction based
92. Powers ET, Morimoto RI, Dillin A, Kelly JW, Balch WE. Bio- on protein fragment complementation assay. Curr Protein Pept
logical and chemical approaches to diseases of proteostasis defi- Sci. 2020;21(6):598–610.
ciency. Annu Rev Biochem. 2009;78:959–91.

13
Pharmaceutical Research

108. Bannister TD, Wang H, Abdul-Hay SO, Masson A, Madoux F, 111. Cuthbertson CR, Guo H, Kyani A, Madak JT, Arabzada Z, Nea-
Ferguson J, Mercer BA, Schurer S, Zuhl A, Cravatt BF, Leis- mati N. The dihydroorotate dehydrogenase inhibitor Brequinar is
sring MA, Hodder P. ML345, a small-molecule inhibitor of the synergistic with ENT1/2 inhibitors. ACS Pharmacol Transl Sci.
insulin-degrading enzyme (IDE). In: Probe reports from the NIH 2020;3(6):1242–52.
molecular libraries program. Bethesda: National Center for Bio- 112. Allotey-Babington GL, Amponsah SK, Nettey T, Sasu C, Nettey
technology Information (US); 2010. H. Quinine Sulphate microparticles as treatment for Leishmania-
109. Escudero MA, Vilanova E. Purification and characterization sis. J Trop Med. 2020;2020:5278518.
of naturally soluble neuropathy target esterase from chicken
sciatic nerve by HPLC and western blot. J Neurochem. Publisher’s Note Springer Nature remains neutral with regard to
1997;69(5):1975–82. jurisdictional claims in published maps and institutional affiliations.
110. Lindner T, Loktev A, Altmann A, Giesel F, Kratochwil C, Debus
J, Jäger D, Mier W, Haberkorn U. Development of Quinoline-
based Theranostic ligands for the targeting of fibroblast activa-
tion protein. J Nucl Med. 2018;59(9):1415–22.

13

View publication stats

You might also like