Skip to main content

The potential role of human islet amyloid polypeptide in type 2 diabetes mellitus and Alzheimer’s diseases

Abstract

Human Islet amyloid polypeptide (hIAPP) from pancreatic β cells in the islet of Langerhans has different physiological functions including inhibiting the release of insulin and glucagon. Type 2 diabetes mellitus (T2DM) is an endocrine disorder due to relative insulin insufficiency and insulin resistance (IR) is associated with increased circulating hIAPP. Remarkably, hIAPP has structural similarity with amyloid beta (Aβ) and can engage in the pathogenesis of T2DM and Alzheimer’s disease (AD). Therefore, the present review aimed to elucidate how hIAPP acts as a link between T2DM and AD. IR, aging and low β cell mass increase expression of hIAPP which binds cell membrane leading to the aberrant release of Ca2+ and activation of the proteolytic enzymes leading to a series of events causing loss of β cells. Peripheral hIAPP plays a major role in the pathogenesis of AD, and high circulating hIAPP level increase AD risk in T2DM patients. However, there is no hard evidence for the role of brain-derived hIAPP in the pathogenesis of AD. Nevertheless, oxidative stress, mitochondrial dysfunction, chaperon-mediated autophagy, heparan sulfate proteoglycan (HSPG), immune response, and zinc homeostasis in T2DM could be the possible mechanisms for the induction of the aggregation of hIAPP which increase AD risk. In conclusion, increasing hIAPP circulating levels in T2DM patients predispose them to the development and progression of AD. Dipeptidyl peptidase 4 (DPP4) inhibitors and glucagon-like peptide-1 (GLP-1) agonists attenuate AD in T2DM by inhibiting expression and deposition of hIAP.

Introduction

Human Islet amyloid polypeptide (hIAPP) also called amylin is one of the main secretory products of pancreatic β cells in the islet of Langerhans [1]. hIAPP has different physiological functions including inhibiting gastric emptying, regulation of satiety and inhibiting the release of insulin and glucagon [1]. In addition, hIAPP has other effects including inhibition of bone resorption, vasodilation and regulation of the renin-angiotensin system (RAS) (Fig. 1) [1, 2].

Fig. 1
figure 1

The physiological role of human Islet amyloid polypeptide (amylin)

hIAPP is co-secreted with insulin from pancreatic β cells, and due to its ability to aggregate into pancreatic islet cells in type 2 diabetes mellitus (T2DM) so-called hIAPP [2]. Aggregation of hIAPP induces cytotoxic effects leading to the loss of pancreatic β cells by inducing apoptosis with subsequent failure of pancreatic β cells and development of T2DM [3].

Notably, hIAPP was initially described in 1901 by weichselbam et al. and Opie et al. as an islet amyloid called islet hyalinization in patients with T2DM [4]. However, islet hyalinization is not specific to T2DM as it is present even in non-diabetic patients [5]. Cohen and Calkins [6] described the fibrous components of different amyloid and islet hyalinization. From these studies, it was concluded that islet hyalinization is part of amyloid proteins. Later on, in 1972 Pearse and his colleagues believed that islet amyloid fibrils might be a product of insulin or proinsulin [7]. In 1986, Westermark et al. described a novel peptide related to the calcitonin gene-related peptide (CGRP) in the pancreatic β cells extracted from insulinoma which was named an insulinoma amyloid peptide [8]. This peptide was renamed as a diabetes-associated peptide in 1987 as hIAPP or amylin later on [9, 10]. Of note, hIAPP is a 37 amino acid hormone co-released and co-stored with insulin and shared a similar enzymatic process with other hormones like CGRP, calcitonin and adrenomedullin [11]. It has been shown that hIAPP is released in a pulsatile manner similar to insulin and circulated in glycosylated and non-glycosylated forms. In healthy subjects, hIAPP circulated levels ranged from 4-25pmol/l, distributed equally with insulin [1, 12]. Unlike insulin which is eliminated by the liver, hIAPP is eliminated by renal metabolism [1]. As well, unlike the insulin gene which is located on chromosome 11, the hIAPP gene is located on chromosome 12 [13]. Pre-pro-hIAPP in the endoplasmic reticulum is converted to pro-hIAPP in the secretory vesicles [14]. Both pro-hIAPP and pro-insulin are processed by endoproteases in the Golgi and secretory granules in a PH-dependent manner [1].

Expression of hIAPP is not limited to the pancreatic β cells but is also expressed on α cells of the islet [11]. In addition, hIAPP is expressed in sensory neurons, the large intestine, the antrum, the brain, and the pancreas [11]. It has been shown that proinsulin and insulin prevent aggregation of hIAPP to form amyloid [15]. Insulin is regarded as a potent inhibitor of hIAPP aggregation, thus insulin resistance (IR) promotes aggregation of hIAPP to form amyloid fibril in the pancreatic β cells [16]. Since, hIAPP is co-released with insulin, though plasma concentration of hIAPP represents 1–2% of that of insulin [16, 17]. Following glucose stimulation, plasma hIAPP is stimulated and increased in parallel with that of insulin [17]. hIAPP is mainly metabolized by the insulin-degrading enzyme (IDE) and neprilysin (NEP) [18, 19]. IDE is a Zinc-metalloprotease expressed in different cells involved in the degradation of hIAPP and amyloid beta (Aβ) preventing hIAPP and Aβ-induced cytotoxicity [18]. NEP is a Zinc-metalloprotease degraded both of hIAPP and Aβ [18, 20]. NEP is highly expressed in pancreatic β cells however; its expression is reduced by aging [18]. Of interest, hIAPP acts on the specific receptors called receptor activity modifying proteins (RAMPs) which are expressed in the brain and renal cortex [20]. Remarkably, hIAPP has structural similarity with Aβ and can involve in the pathogenesis of T2DM [21] and AD [22]. Therefore, the present review aimed to elucidate how hIAPP acts as a link between T2DM and AD.

Role of hIAPP in T2DM

T2DM is an endocrine disorder due to relative insulin insufficiency and IR, characterized by hyperglycemia and cardiometabolic complications [22]. T2DM represents 95% of all diabetes types and contributes to 5% of all mortalities under the age of 70 [23, 24]. Genetic, epigenetic, environmental factors, obesity, smoking and male sex are the main risk factors involved in the development and progression of T2DM [25, 26]. Particularly, aggregation of hIAPP is associated with the development of T2DM by inducing apoptosis and loss of pancreatic β cells in the islet of Langerhans [21]. However, hIAPP had been reported to have a protective effect against the degeneration of pancreatic β cells in mice with alloxan-induced diabetes [27]. The protective effect of hIAPP on the pancreatic β cells is through the improvement of pancreatic microcirculation and limit hyperpolarization of β cells [27]. A case-control study involved diabetic patients and healthy controls showed that plasma hIAPP was lower than insulin in healthy subjects; it increased in obese subjects and was undetectable in T1DM. In T2DM obese patients, plasma hIAPP level was similar to healthy controls but low in T2DM non-obese patients, suggesting the role of hIAPP with insulin in the regulation of glucose homeostasis [28].

Deficiency of IDE which is involved in the degradation of hIAPP induces the development of hyperamylinemia which triggers the development of IR [29]. In vitro study demonstrated that inhibition of IDE increases the risk of hIAPP-induced cytotoxicity [30]. However, acute administration of IDE inhibitor leads to beneficial effects on glucose tolerance in mice by increasing hIAPP which decreases gastric empty time [31]. Different studies illustrated that chronic use of IDE inhibitors leads to metabolic dysfunction via dysregulation of the proteasome pathway and chaperon-mediated autophagy [30, 32]. In vitro study demonstrated that inhibition of IDE by bacitracin increases the risk for the development of hIAPP-induced cytotoxicity through inhibition clearance of hIAPP [30]. In contrast, inhibition of IDE may improve glucose homeostasis [32].

Notably, hIAPP unlike rat IAPP is highly subjected to amyloidogenesis by forming amyloid fibril. The amyloidogenic propensity of hIAPP is related to the presence of 20–29 and 8–18 residues which have no proline as in rat IAPP as proline prevents aggregation of hIAPP [33]. Higher level of plasma hIAPP in prediabetic and diabetic patients promotes the formation of hIAPP protofibrils leading to progressive dysfunction of pancreatic β cells with the development of T2DM which in turn promote the secretion of hIAPP in a vicious cycle (Fig. 2) [33].

Fig. 2
figure 2

Human Islet amyloid polypeptide (hIAPP) and type 2 diabetes mellitus (T2DM)

In addition, a monoclonal antibody to hIAPP could be an early diagnostic biomarker for T2DM diagnosis [33]. These findings raised an intricate question is hIAPP represents dysfunction of pancreatic β cells or is it an etiological factor in the development of T2DM? Clark et al. [34] suggested that the deposition of hIAPP and formation of pancreatic β cells amyloidosis is a consequence of T2DM since in humans T2DM islet amyloidosis is present in less than 1% to more than 80%. Extensive deposition of hIAPP is correlated with the reduction of dysfunction of pancreatic β cells in animal and human studies [35, 36]. Islet amyloidosis is not developed in T1DM where both insulin and hIAPP are absent [34]. Bram et al. [37] observed that hIAPP-induced pancreatic β cell apoptosis was prevented by diabetes-associated antibodies. However, glucose intolerance and hyperglycemia promote oligomerization of hIAPP [34]. Once protofibril is formed, it acts as nucleation nidus promoting progressive fibril generation and development of IR [34]. Remarkably, there is a vicious cycle between hIAPP and glycated insulin through exacerbation of pancreatic β cytotoxicity in T2DM [38]. Notoriously, sulfonylurea promotes fibril formations in diabetic cats [39]. ATP-sensitive potassium channel, a site activated by sulfonylurea, is required for hIAPP to induce pancreatic β cells dysfunction [40, 41].

Therefore, the oligomerization of hIAPP is controlled by the balance between processing and degradation pathways [42]. Genetic variants of these pathways affect the prevalence of T2DM in the Chines population [42]. Other factors are also involved in enhancing the oligomerization of hIAPP to amyloid fibrils and the formation of amyloid in the pancreatic β cells [43]. Not all forms of hIAPP aggregates induce the destruction and apoptosis of pancreatic β cells, though only toxic hIAPP aggregates can cause these events [43].

The underlying mechanisms that increase hIAPP aggregates

It has been shown that the incidence and prevalence of T2DM are increased with age due to increase aggregation of hIAPP and the development of pancreatic β cells [44]. Of interest, hIAPP and associated monomers have normal biological activity on normal pancreatic β cells [45]. However, toxic oligomers lead to membrane toxicity in T2DM [45]. Toxic non-soluble fibrils from hIAPP interact with lipids in the cell membrane disrupting negative charge phospholipid [45]. Besides, the hydrophobic part of non-soluble fibrils induces protein misfolding at acidic PH [46].

The underlying causative factors that convert soluble hIAPP oligomers to toxic non-soluble fibrils are largely unknown [47]. Nevertheless, different environmental factors may induce mutations like the S20G mutation which result in more formation of the toxic non-soluble fibrils [47]. As well, a single mutation in F15 of hIAPP oligomers increases the biosynthesis of toxic non-soluble fibrils [48]. These mutations affect the solubility of hIAPP oligomers by changing the sequence of proline [49]. In addition, abnormal processing of hIAPP may promote the formation of toxic non-soluble fibrils [48, 49]. The absence of proprotein convertase 2 which involves the processing and removal of hIAPP oligomers enhances the generation of toxic non-soluble fibrils and induction dysfunction of pancreatic β cells [49]. In addition, the dysfunction of the deamidation process which is involved in the stability, structure, and aggregation of hIAPP oligomers by adding a negative charge promotes the production of unmodified hIAPP oligomers and induces the formation of toxic non-soluble fibrils [50]. Abnormality in the disulfide bonds C2-C7 increases the formation of toxic non-soluble fibrils by affecting the kinetic process of hIAPP oligomers [51]. It has been shown that copper ions encourage the aggregation of hIAPP oligomers by inducing reactive oxygen species (ROS) [52]. Copper chelating agents can reduce T2DM risk by reducing the formation of ROS and the production of unmodified hIAPP oligomers [52]. Notoriously, heparin plays a critical role in the production of hIAPP oligomers and the formation of toxic non-soluble fibrils [53]. The negative charge of heparin binds the positive charge in the N-terminal of hIAPP oligomers. This binding enhances hIAPP oligomers to be converted to toxic non-soluble fibrils and attenuates hIAPP-induced cytotoxicity [53].

Mechanisms of hIAPP oligomers-induced cytotoxicity

Monomeric and oligomeric hIAPP disrupt cell membrane fluidity by increasing the production of ROS causing membrane leakage and cytotoxicity [54]. Monomeric hIAPP can interact with different proteins leading to cytotoxicity [54]. A higher concentration of hIAPP is subjected to form aggregation [55]. For example, the higher concentration of hIAPP in IR enhances its aggregation with the formation of toxic non-soluble fibrils [55]. Furthermore, the higher concentrations of hIAPP and aggregated fibrils can induce endoplasmic reticulum (ER) stress which are the hallmark of T2DM [56]. In turn, oxidative stress, ER stress, and hyperglycemia promote the aggregation of toxic non-soluble fibrils [56]. Markedly, IR, aging, and low β cell mass increase expression of hIAPP which binds cell membrane leading to the aberrant release of Ca2+ and activation of proteolytic enzyme calpain leading to a series of events causing loss of β cells [57] (Fig. 3).

Fig. 3
figure 3

Human Islet amyloid polypeptide (hIAPP) and loss of β cells

In response to oxidative stress and ER stress, chaperon-mediated autophagy is upregulated to prevent cell injury [58]. Chaperon-mediated autophagy can bind hIAPP and prevent its aggregation [58]. It has been shown that impairment of autophagy contributes to the accumulation of toxic non-soluble fibrils and the development of dysfunction of pancreatic β cells [58]. Therefore, restoration of autophagy and proteostasis of hIAPP may prevent hIAPP-induced cytotoxicity. In T2DM patients, the serum level of chaperone heat shock protein 70 (Hsp70) is increased [59]. A case-control study included 36 newly diagnosed T2DM patients, 37 long-standing T2DM patients and 36 healthy controls showed that chaperon Hsp70 serum level was higher in T2DM patients and correlated with the disease duration compared to the controls [59]. Ladjimi et al. [60] observed that chaperon Hsp70 in T2DM is augmented to inhibit aggregation of hIAPP. Therefore, increased chaperone Hsp70 serum level could be a compensatory mechanism against the development of oxidative stress and ER stress in T2DM. Augmentation of endogenous or use of exogenous chaperon Hsp70 activators could be effective in the attenuation of hIAPP-mediated pancreatic β cytotoxicity [60]. Administration of alfalfa-derived Hsp70 in patients with IR could be effective in the prevention of T2DM [60].

Furthermore, heparan sulfate proteoglycan (HSPG) can increase hIAPP aggregation [61]. Perlecan is a secretory part of HSPG that can bind hIAPP and increase its aggregation and cytotoxicity as confirmed in vitro and in vivo [61]. Loss of perlecan in transgenic mice prevents the accumulation of hIAPP and maintains glucose homeostasis [61]. In the components of islet amyloid aggregates, there is also serum amyloid protein, apolipoprotein E4 and HSPG which increase the aggregation of hIAPP [61]. Perlecan which is localized in the pericapillary matrix of the islet is mainly involved in the aggregation of hIAPP [61].

Moreover, the immune response to the formed hIAPP fibrils triggers the release of pro-inflammatory cytokines mainly IL-1β which increases hIAPP fibrils-induced β cytotoxicity and development of IR and T2DM [62]. In vitro study demonstrated that hIAPP activates toll-like receptor 2 (TLR2) with activation expression of nuclear factor kappa B (NF-κB) and induces the expression of pro-inflammatory cytokines [63]. This finding suggests a critical role of pro-inflammatory cytokines in the development and progression of inflammation in the pancreatic islet with the development of IR. Westwell-Westwell-Roper et al. [64] found that blocking IL-1β can attenuate hIAPP-induced cytotoxicity and pancreatic β cell dysfunction. The host defense mechanism against hIAPP induces the release of immunomodulatory cathelicidin which suppress the assembly of hIAPP and the associated injury of pancreatic β cell [65]. Thus, the immune response against hIAPP has bidirectional effects that could be harmful or beneficial.

Indeed, zinc prevents protein misfolding and aggregation of hIAPP [66]. Zinc deficiency enhances the aggregation of hIAPP and the development of injury of pancreatic β cells [66]. In vitro study demonstrated that zinc can bind un-aggregated hIAPP at a micro-microlar concentration [66]. Of interest, the mutation of zinc ion transporter on the secretory granules reduces hIAPP aggregation [67]. Brender et al. [68] illustrated that zinc has an inhibitory effect on the aggregation of hIAPP by increasing lag-time for hIAPP aggregation and interrupting the formation of toxic non-soluble fibrils. Zinc level is reduced in T2DM patients compared to healthy controls as documented in a case-control study [68]. Zinc deficiency in T2DM patients is associated with poor glycemic control [69] and this may explain why zinc deficiency enhances hIAPP aggregation and the associated dysfunction of pancreatic β cells.

In sum, hIAPP-induced dysfunction of pancreatic β cells through induction of oxidative stress and ER stress. Zinc deficiency and exaggerated immune response against hIAPP aggregation trigger more injury of pancreatic β cells.

Role of hIAPP in AD

AD is the most common type of dementia characterized by progressive neurodegeneration leading to memory and cognitive deficits [70]. The pathological hallmarks of AD neuropathology are the deposition of extracellular Aβ and intracellular aggregation of tau protein which form neurofibrillary tangles (NFTs) [71]. AD affects more than 50 million individuals worldwide. One-third of AD cases are attributed to modifiable risk factors including obesity and T2DM [72]. It has been reported that the development of AD is linked to the duration and severity of T2DM [73].

T2DM patients have been reported to hold a higher incidence of cognitive decline and AD; T2DM has been intensely associated with an increased risk of developing all types of dementia, including AD [74]. A systematic review including 14 longitudinal population-based studies found that the incidence of any dementia was higher in T2DM patients than in those without T2DM [75]. Some studies have relied on the self-reported diagnosis of T2DM, and in the elderly population, many patients with T2DM may remain undiagnosed [76]. In a longitudinal cohort study, lasting up to 9 years, the risk of developing AD was 65% higher in T2DM patients as compared to the non-diabetic controls [77]. In a community-based controlled study, the prevalence of T2DM and glucose intolerance was examined in patients with AD vs. control participants without AD. The study suggested that T2DM was 35% and glucose intolerance was 46% might be present in up to 80% of patients with AD [78]. It has been shown that the incidence of dementia was higher in subjects with diabetes (14.9%) than that in nondiabetic subjects (10.3%) during the examination period between 1992 and 1999, with hazard ratio of 1.62 for AD in subjects with T2DM [78].

Disturbances in brain insulin signaling mechanisms represent early and progressive abnormalities and could account for the majority of molecular, biochemical, and histopathological lesions in AD [79]. Increasing IR and hyperinsulinemia were linked with more hippocampal and amygdale atrophy on magnetic resonance imaging in T2DM patients when compared to matched controls, regardless of vascular pathology [80]. It has been suggested that may be a common underlying mechanism predisposes to amyloid deposition in the brain and in the pancreatic islet [78].

Amyloid formation is a hallmark in both AD and T2DM due to the deposition of hIAPP in both pancreatic β islets and the brain [61]. Pancreatic-derived hIAPP can cross the blood-brain barrier (BBB). Besides, brain-derived hIAPP can interact with Aβ leading to progressive neuronal injury [81]. Brain and pancreatic hIAPP increase Aβ misfolding which are involved in AD neuropathology [82, 83]. Aggregation of hIAPP a pathological hallmark of T2DM is also observed in AD. Both hIAPP and Aβ exert similar cytotoxic mechanisms and share common physiochemical properties [83]. Similar to Aβ deposition in AD, hIAPP also aggregates in patients with T2DM to form pancreatic islet amyloid. Further, hIAPP and Aβ have a total of 25% amino acid sequence identity with high binding affinity to each other. They also share many biophysical and physiological properties and exert similar cytotoxic mechanisms when they aggregate. Furthermore, IAPP deposits have been found in the brain tissue of patients with AD, contributing to the pathophysiology of the disease [83]. These facts provide the basis to hypothesize that aggregation of amyloidogenic IAPP during T2DM plays a key role in the pathogenesis of AD [84]. The cross-seeding Aβ–hIAPP assemblies showed a wide range of polymorphic structures via a combination of four β-sheet-to-β-sheet interfaces and two packing orientations, focusing on a comparison of different matches of β-sheet layers [83]. Two cross-seeding Aβ–hIAPP assemblies with different interfacial β-sheet packings exhibited high structural stability and favorable interfacial interactions in both oligomeric and fibrillar states [83]. Both Aβ–hIAPP assemblies displayed interfacial dehydration to different extents, which in turn promoted Aβ–hIAPP association depending on interfacial polarity and geometry [84, 85]. Inflammation in the brain and pancreas is associated with cell degeneration and pathogenesis of both AD and T2DM. Inflammatory cascades in both tissues are triggered by the uptake of Aβ or IAPP aggregates by microglial cells or macrophages and their insufficient lysosomal degradation. This results in lysosomal damage, caspase-1/node like receptor pyrin 3 (NLRP3) inflammasome activation and release of IL-1β, a key pro-inflammatory cytokine in both diseases [83]. The inflammatory processes mediated by Aβ and IAPP aggregates are blocked by IAPP-GI, a non-amyloidogenic IAPP mimic, which forms high-affinity soluble and nonfibrillar hetero-oligomers with both polypeptides [83]. In contrast to fibrillar Aβ aggregates, nonfibrillar Aβ/IAPP-GI or Aβ/IAPP hetero-oligomers become rapidly internalized by microglial cells and targeted to lysosomes where Aβ is fully degraded. Internalization occurs via IAPP receptor-mediated endocytosis. In contrast to IAPP aggregates, IAPP/IAPP-GI hetero-oligomers become rapidly internalized and degraded in the lysosomal compartments of macrophages. Therefore, IAPP/Aβ cross-amyloid interaction and suggest that conversion of Aβ or IAPP into lysosome-targeted and easily degradable hetero-oligomers by hetero-association with IAPP mimics could become a promising approach to specifically prevent amyloid-mediated inflammation in AD, T2D, or both diseases [83].

However, there was no correlation between peripheral hIAPP and AD risk in animal and human studies [83]. In the Tg2576 AD mouse model, IAPP plasma levels were not significantly elevated at an age where the mice exhibit the glucose intolerance of pre-diabetes. Based on this negative data, it appears unlikely that peripheral IAPP cross-seeds Aβ pathology in the AD brain. However, IAPP protein is present in astrocytes in the murine brains and secreted from primary cultured astrocytes [83]. This preliminary report suggests a potential and novel association between brain-derived IAPP and AD, however, whether astrocytic-derived IAPP cross-seeds Aβ in the brain necessitates additional research.

Peripheral hIAPP serum level is not different in AD and controls [83]. Therefore, peripheral hIAPP may not be the partner in AD neuropathology, though brain-derived hIAPP could be the main in AD neuropathology. However, cerebrospinal fluid (CSF) hIAPP level is augmented in AD patients with or without T2DM [83]. Moreover, deposition of hIAPP alone or mixed with hIAPP in the temporal lobe are increased in both T2DM and late-onset AD patients [86]. High hIAPP in AD implicates its role in AD neuropathology, however, the origin of hIAPP could be from the brain, or pancreatic islet remain not fully elucidated [87]. An experimental study confirmed that local brain production of hIAPP contributes to AD neuropathology [87]. Therefore, hIAPP was detected in both brain parenchyma and CSF of AD patients [88].

Deposition of hIAPP in both T2DM and AD is due to metabolic dysfunction, microvascular injury, and failure of Aβ clearance [86]. Besides, the deposition of tau proteins and Aβ are also found in the pancreatic islet [89]. These verdicts indicate the association between T2DM and AD (Fig. 4).

Fig. 4
figure 4

Role of human Islet amyloid polypeptide (hIAPP) in Alzheimer’s disease (AD)

In the normal state, unmodified (soluble) hIAPP crosses BBB and enhances clearance of Aβ4 to prevent the development and progression of AD neuropathology [90]. When unmodified hIAPP is subjected to oxidative stress factors, it becomes modified with subsequent loss of its protective biology. Modified hIAPP has the propensity to aggregate and be involved in AD neuropathology [90]. Schultz et al. [90] elucidated that total CSF hIAPP level was not differed in AD compared to individuals with cognitive impairments. However, total CSF hIAPP level was positively correlated with tau protein level and negatively correlated with CSF Aβ42 level [90]. Both hIAPP and Aβ have interacted and seeded each other to form hybrid amyloid fibrils [85]. The correlation between plasma and CSF levels of unmodified hIAPP suggests a well-functional translocation of peripherally-derived unmodified hIAPP over the BBB under normal conditions. This crossing is important for brain processes dependent on hIAPP access, such as appetite regulation and cognition [83]. Interestingly, the correlation between CSF and plasma unmodified hIAPP levels was lost in patients with AD both with and without T2DM; a result indicating either that normal BBB crossing of unmodified hIAPP is compromised or clearance of unmodified hIAPP into the CSF is dysfunctional in these patients [91]. In contrast to unmodified hIAPP, levels of total IAPP in plasma did not correlate with the same in CSF. This disassociation could be due to a limitation of the amount of IAPP that can cross the BBB [83, 91]. In particular, hIAPP is associated with a higher AD risk by different mechanisms. Circulating hIAPP affects neuronal functions independent of Aβ42 level even in non-diabetic status, hIAPP disrupts neurovascular injury, interacts with Aβ42 and acts as a seed to increase deposition of Aβ plaques [92]. In addition, hIAPP exacerbates the toxic effects of Aβ42 by increasing the production of ROS. Likewise, Aβ42 interacts, cross seed and increases the toxic effects of hIAPP [93]. Co-aggregation of hIAPP and Aβ42 increases tau protein hyperphosphorylation through induction of oxidative stress and inflammatory changes causing progressive synaptic loss and synaptic dysfunction [94]. IAPP colocalizes with pathological tau in AD brain. IAPP binds tau and promotes its aggregation into a more toxic strain that shows increased seeding activity and neurotoxicity in vitro. Intra-hippocampal injection of the IAPP-modified tau fibrils into the tau transgenic mice induced more severe tau pathology and cognitive deficits when compared with tau fibrils. Hence, our results indicate that IAPP cross-seeds tau and mediates the spreading of tau pathology in AD [94, 95]. Synthetic Aβ aggregates intravenously injected into hIAPP transgenic mice triggered IAPP amyloid formation, and accelerated pancreatic pathology, supporting the cross-seeding interaction between these peptides [87]. Given that both hIAPP and tau are amyloidogenic and prone to aggregate into amyloid deposits, hIAPP may interact with tau and enhance tau pathology [88, 96]. However, the factors that initiate the formation of different strains remain elusive.

A recent study involved postmortem brain tissues of AD patients together with in vitro and experimental studies illustrated that hIAPP promotes tau protein deposition [95]. In addition, peripheral hIAPP promotes the conversion of tau protein to more toxic tau fibrils [94] suggesting that hIAPP plays an assisting role in AD neuropathology. However, the assembly of Aβ42 is prevented by hIAPP and hIAPP analogue pramlintide which attenuate the development of AD and other neurodegenerative diseases [97]. Taken together, peripheral hIAPP plays a major role in the pathogenesis of AD, and high circulating hIAPP level increase AD risk in T2DM patients. However, there is no hard evidence for the role of brain-derived hIAPP in the pathogenesis of AD.

Discussion

Interestingly, hIAPP undergoes tertiary structural changes and can form aggregates that deposit in the pancreatic islet causing β cells dysfunction even in the prediabetic period [98]. Both AD and T2DM share a common feature of protein deposits, hIAPP in TDM and Aβ in AD. In addition, hIAPP shares many physiochemical properties of Aβ [98]. Particularly, both Aβ and hIAPP are degraded by IDE and NEP, therefore mutation of these enzymes prevents degradation of Aβ and hIAPP with subsequent accumulation in both brain and pancreatic islets leading to AD and IR [99]. Therefore, boosting the activity of IDE and NEP may reduce the risk for the development of both AD and T2DM. The potential role of hIAPP in the development and progression of AD in T2DM was well elucidated by different studies [100, 101]. However, the novelty of the present review is to review the mechanistic role of pancreatic and brain-derived hIAPP in the induction of AD neuropathology.

The mode of toxicity by hIAPP and Aβ despite is mediated in a receptor-dependent manner. Receptor activity modifying protein 3 (RAMP3) and calcitonin receptors which are highly expressed in the brain act as a receptor for hIAPP to increase Aβ toxicity [102]. In addition, the expression of hIAPP receptors is correlated with Aβ load since these receptors are required for the effect of Aβ [102]. In this state, Aβ-induced cholinergic toxicity in rats is prevented by hIAPP antagonists which also attenuate Aβ-mediate apoptosis [103]. Aβ and hIAPP induced neuronal injury and neurotoxicity through the activation of integrin signaling [104]. Likewise, Aβ and hIAPP can increase Aβ and associated AD neuropathology by augmentation of the expression of amyloid precursor protein (APP) [105]. Therefore, Aβ neurotoxicity is mediated by the action of hIAPP. Comparable to Aβ, hIAPP can cause apoptosis and neuronal cell deaths as confirmed by in vitro study [102]. Aβ-hIAPP oligomerized mixture has three-fold more neurotoxic effects compared to hIAPP and Aβ alone [102]. In particular, Aβ-hIAPP oligomerized, hIAPP and Aβ lead to more neurotoxic effects on the hippocampal neurons [102] indicating the vulnerability of hippocampal neurons to the neurotoxic effect of hIAPP and Aβ. Notably, hIAPP and Aβ can bind neuronal and non-neuronal cell membranes leading to synaptic dysfunction through the induction of oxidative stress and mitochondrial dysfunction [106]. These verdicts proposed that hIAPP and Aβ can interact at different levels to induce neurotoxicity and neuronal injury to induce the development and progression of AD. However, not all T2DM patients develop AD in relation to high circulating hIAPP. Thus, precipitating factors in T2DM and AD could be the underlying mechanisms for the aggregation of hIAPP and increase the interaction between hIAPP and Aβ. Of note, oxidative stress and mitochondrial dysfunction are common in both T2DM and AD [56, 107]. These pathological conditions enhance aggregation and promote the interaction of hIAPP with Aβ to induce neurotoxicity [108]. As mentioned above, chaperon-mediated autophagy binds and increases the clearance of hIAPP [58] this process is dysregulated in both T2DM and AD [58, 109]. Therefore, cellular dysfunctions including oxidative stress, mitochondrial dysfunction, and dysregulated chaperon-mediated autophagy could the possible mechanism for the aggregation of hIAPP with the development of AD neuropathology in T2DM. The use of antioxidants or autophagy activators may reduce AD risk in T2DM patients [109, 110].

Interestingly, HSPG and perlecan which increase hIAPP aggregation [61] are dysregulated in T2DM and associated complications [111]. Hyperglycemia in T2DM induces significant alteration of HSPG in the endothelium and brain by increasing expression of heparanase enzyme which degrades HSPG [112]. Dysregulated HSPG in T2DM promotes neuronal injury by increasing the aggregation of hIAPP [61] leading to the development and progression of AD. It has been shown that HSPG is highly expressed in the brain mainly in Aβ deposits in transgenic mice and AD patients [113]. In AD, the expression of HSPG is altered in the brain [114]. A case-controlled study on 18 AD patients and 6 healthy controls revealed that core protein HSPG was increased in AD patients compared to the control [115]. This finding suggests that HSPG could be a potent inducer of AD neuropathology through the increasing aggregation of hIAPP. Thus, dysregulated HSPG in T2DM patients enhances hIAPP aggregation leading to the development of AD.

Furthermore, abnormal immune response to the aggregated hIAPP triggers releases of pro-inflammatory cytokines and expression of inflammatory signaling pathways that augment hIAPP-induced cytotoxicity in both T2DM [62] and AD [115]. Of note, NF-κB is involved in the pathogenesis of AD and T2DM leading to neuroinflammation and inflammatory reaction in the pancreatic islet respectively [116, 117]. NF-κB accelerates aggregation of hIAPP in mice [63]. Likewise, the NLRP3 inflammasome which is activated in both AD and T2DM [116, 117] also accelerates the aggregation of hIAPP [118]. These findings indicated that an exaggerated immune response against hIAPP induces more pancreatic islet cytotoxicity and neurotoxicity.

In addition, zinc has a protective effect against protein misfolding and aggregation of hIAPP [66]. Zinc deficiency enhances the aggregation of hIAPP and the development of injury of pancreatic β cells [66]. It has been reported that zinc level is reduced in both T2DM and AD [69, 119]. Zinc has a neuroprotective effect in normal concentration, though a higher concentration of zinc leads to neurotoxicity [119]. Zinc modulates APP function and inhibits protein phosphatase 2 A which is involved in tau protein phosphorylation [12]. However, higher zinc level promotes tau protein hyperphosphorylation and AD neuropathology [119]. Therefore, zinc has bidirectional effects that could be beneficial or detrimental to AD neuropathology. In this state, zinc dyshomeostasis in T2DM affects AD neuropathology through modulation of the aggregation of hIAPP.

Taken together, oxidative stress, mitochondrial dysfunction, chaperon-mediated autophagy, HSPG, immune response, and zinc homeostasis in T2DM could be the possible mechanisms for induction of the aggregation of hIAPP which increase AD risk.

Effects of anti-diabetic drugs on hIAPP and AD

Insulin sensitizing agents

It has been detected that IR is linked with mild cognitive impairment in AD patients compared to controls due to dysfunction of brain insulin signaling [120]. Administration of insulin did not reduce IR in postmortem brain tissue [120]. Consequently, overcoming IR by insulin-sensitizing agents could be more effective in reducing brain IR and AD neuropathology. Metformin is an insulin-sensitizing agent that belongs to the biguanid group used as first-line therapy in the management of T2DM and has the capability to decrease peripheral IR [121]. Metformin can reduce the production and aggregation of Aβ in the hippocampus and cerebral cortex by increasing the activity of IDE [122]. AD is regarded as type 3 diabetes mellitus (T3DM) due to the sharing similarity with T2DM and the development of brain IR [123]. Therefore, metformin may act in a double role in the prevention and treatment of both T2DM and AD. Preclinical and pilot studies propose that metformin therapy is effective in the management of AD [124]. Treatment of mice with metformin for 8 weeks amended cognitive function by inhibiting Aβ accumulation [124]. A randomized study exemplified that metformin recovers memory and cognitive functions [125]. Additionally, a modern study confirms that metformin advances cognitive and memory function reduces Aβ accumulation and links to inflammation and oxidative stress [126]. An epidemiological study on T2DM patients on diverse diabetic treatments demonstrated that long-term metformin therapy was linked with more reduction in the hazard ratio for development of AD compared to sulfonylurea [127]. As well, long-standing metformin therapy improves learning function and attention ability by increasing orbito-frontal neuronal metabolism which extremely concerned in AD [128]. This finding suggests that metformin leads to a selective effect on brain regions. Interestingly, metformin has ability to cross BBB and controls brain IR by modulating the expression of adenosine monophosphate kinase protein kinase (AMPK) which augments expression of insulin receptor substrate 1 (IRS-1) [129] leading to the inhibition the production of Aβ, tau protein phosphorylation and induction of autophagy [130]. Likewise, metformin attenuates IR development and reduces the expression of tau hyperphosphorylation [131]. Brain IR triggers the expression of tau protein kinases like glycogen synthase kinase 3 β (GSK3β) which increase tau hyperphosphorylation, misfolding and oligomerization in postsynaptic neurons [132]. IDE and GSK3β are altered in T2DM and AD. Upregulation of GSK3β and downregulation of IDE are associated with the development of AD in T2DM [133]. Brain IR may develop due to Aβ accumulation as deletion of Aβ enhances peripheral and brain insulin sensitivity [134]. These findings proposed a vicious cycle between Aβ accumulation and brain IR. Besides, chronic hyperglycemia and associated glucolipotoxicity in T2DM promote the generation of advanced glycation end-products (AGEs) which are regarded as a possible link between T2DM and late-onset AD [135]. Increasing of brain AGEs promote development of AD through activation of APP processing, deposition of Aβ and Aβ fibrilisation. Likewise, AGEs trigger the expression of AGEs receptors which are considered as possible receptors of Aβ [136]. Metformin regulates Aβ production and associated oxidative stress and mitochondrial dysfunction. As well, metformin reduces apoptotic cell deaths and increases neurogenesis [137]. The beneficial effects of metformin therapy in AD are by different mechanism including normalization of tau protein metabolism and phosphorylation, regulation of autophagy and Aβ accumulation. In virtue of its anti-inflammatory effect of metformin improves cognitive function and may attenuate the progression of AD neuropathology via induction of Aβ clearance with inhibition of tau protein phosphorylation and associated neuroinflammation [138].

Furthermore, the thiazolidinediones (TZDs), a family of peroxisome proliferator–activated receptor γ (PPARγ) activators like rosiglitazone and pioglitazone are antidiabetic agents that primarily act by improving peripheral insulin sensitivity [139]. TZDs improve pancreatic β-cell function and insulin sensitivity in diabetic animals and T2DM patients [139,140,141].

It has been demonstrated that hIAP formation in vivo is markedly reduced by the effects of rosiglitazone and metformin in hIAPP transgenic male mice fed a moderate-fat diet for 1 year [142]. Rosiglitazone and metformin treatment resulted in reduced β-cell secretory demand, as shown by reduced insulin secretion in response to intravenous glucose, with the maintenance of euglycemia [142]. The mean insulin response in the rosiglitazone-treated mice was unexpectedly low, due to paradoxical negative values for the acute insulin response to glucose in three mice [142]. Thus, in this case, the reduction of islet mass with rosiglitazone and metformin appears to have been appropriate for the agents’ effects to reduce the secretory stimulus to the β-cell [142]. Reduction in the proportion of pancreatic β-cell mass to islet mass was strongly correlated with increased hIAPP severity, consistent with an effect of amyloid to result in β-cell loss [142]. Therefore, treatment with both rosiglitazone and metformin, while resulting in reduced β-cell mass through reduced secretory demand, acted to preserve the remaining β-cell population through suppression of islet amyloid formation. This effect of rosiglitazone and metformin to reduce islet amyloid formation through decreased secretory demand is consistent with studies showing that increased secretory demand due to obesity and IR is linked with marked deposition in hIAPP transgenic mice, whereas decreased insulin output by disrupting β-cell glucokinase resulted in a significant reduction in islet amyloid deposition [143]. Therefore, rosiglitazone or metformin treatment significantly reduces both the prevalence and severity of islet amyloid in hIAPP transgenic mice. These effects appear to be mediated in part through the ability of these agents to alter visceral fat deposition, thus reducing secretory demand on the compromised β-cell that is capable of forming islet amyloid. Further, it would appear that both agents might have an effect beyond simply changing visceral fat, with this effect being greater with rosiglitazone [142, 143]. A systematic review on the effects of insulin sensitizer including metformin and TZDs on AD neuropathology showed these agents were effective in reducing Aβ pathology [144]. Findings from clinical trials and current safety data suggest that rosiglitazone should not be used for the treatment of AD. Application of results from trials evaluating pioglitazone in the treatment of AD is limited because of major trial limitations; therefore, it should not be recommended at this time [145]. However, a preliminary study suggested that rosiglitazone may offer a novel strategy for the treatment of cognitive decline associated with AD [146]. Relative to the placebo group, subjects receiving rosiglitazone exhibited better delayed recall and selective attention. Plasma Aβ levels were unchanged from baseline for subjects receiving rosiglitazone but declined for subjects receiving placebo, consistent with recent reports that plasma Aβ42 decreases with progression of AD [146]. No evidence of the efficacy of 2 mg or 8 mg rosiglitazone monotherapy in cognition or global function was detected in the APOE-Ε4-negative or other analysis populations [147]. These findings proposed that insulin-sensitizing agents’ metformin and TZDs could be effective in both T2DM and AD through modulation of hIAPP.

Dipeptidyl dipeptidase inhibitors and incretin analogues

Dipeptidyl peptidase 4 (DPP4) inhibitors like sitagliptin are widely used and tolerated in the management of T2DM with minimal or no hypoglycemia as an adverse effect [148, 149]. Preclinical studies indicated that DPP4 inhibitors are effective against AD neuropathology [150, 151]. The long-term inhibition of the endogenous DPP-4 enzymes with sitagliptin can meaningfully delay some forms of AD pathology, including Aβ deposition, when administered early in the disease course of a transgenic mouse model of AD [150]. DPP4 inhibitor saxagliptin which increases the level of glucagon-like peptide-1 (GLP-1) and ameliorates T2DM, has become a valuable candidate as a disease-modifying agent in the treatment of AD. In addition, endogenous GLP-1 levels decrease Aβ peptide and tau phosphorylation in the AD rat models [151]. Furthermore, GLP1 receptor agonists have also been shown as possessing neuroprotective effects in AD, which seem to improve nearly all neuropathological features as well as cognitive functions of AD [152]. For example, NFTs, amyloid plaques, and neuro-inflammations in the hippocampus have been reduced in AD model mice [153]. It has been shown that a GLP1 receptor agonist also prevents synaptic damage induced by Aβ accumulation, which supports spatial memory by affecting the phosphoinositide-3 kinase (PI3K)-AKT pathway [154]. Targeting DPP4 inhibitors that is involved in the GLP1 signaling has been considered as a promising therapeutic model for AD [155]. Furthermore, the mammalian/mechanistic target of rapamycin (mTOR) has been considered as a center that integrates multiple signaling cascades including the GLP1 receptor signaling, which may also be involved in the progression of AD [155]. Moreover, treatment with DPP4 inhibitor vildagliptin restored the islet topography by modulating the expression of hIAP [156]. Notably, sitagliptin preserves pancreatic β-cell mass and function and enhances insulin sensitivity in the hIAP rat model of T2DM [157]. Therefore, DPP4 inhibitors and GLP-1 agonists attenuate AD in T2DM by inhibiting expression and deposition of hIAP.

Conclusion

hIAPP is one of the main secretory products of pancreatic β cells in the islet of Langerhans. hIAPP has different physiological functions including inhibiting gastric empty, regulation of satiety and inhibiting the release of insulin and glucagon. T2DM is an endocrine disorder due to relative insulin insufficiency and IR, characterized by hyperglycemia and cardiometabolic complications. Interestingly, hIAPP has structural similarity with Aβ and can involve in the pathogenesis of T2DM and AD. The underlying causative factors that convert soluble hIAPP oligomers to toxic non-soluble fibrils are largely unknown. IR, aging and low β cell mass increases the expression of hIAPP which binds the cell membrane leading to the aberrant release of Ca2+ and activation of the proteolytic enzyme leading to series of events causing loss of β cells. hIAPP-induced dysfunction of pancreatic β cells through induction of oxidative stress and ER stress. Zinc deficiency and exaggerated immune response against hIAPP aggregation trigger more injury of pancreatic β cells. Peripheral hIAPP plays a major role in the pathogenesis of AD, and high circulating hIAPP level increase AD risk in T2DM patients. However, there is no hard evidence for the role of brain-derived hIAPP in the pathogenesis of AD. In sum, oxidative stress, mitochondrial dysfunction, chaperon-mediated autophagy, HSPG, immune response and zinc homeostasis in T2DM could be the possible mechanisms for induction the aggregation of hIAPP which increase AD risk. In addition, DPP4 inhibitors and GLP-1 agonists attenuate AD in T2DM by inhibiting the expression and deposition of hIAP. Despite these evidences the underlying mechanism linking pancreatic islet hIAPP in the development of AD need to be elucidated. Therefore, retrospective and prospective studies are recommended in this state.

Data Availability

Not applicable.

Abbreviations

hIAPP:

Human Islet amyloid polypeptide

T2DM:

Type 2 diabetes mellitus

Aβ:

Amyloid beta

AD:

Alzheimer’s disease

RAS:

Renin-angiotensin system

CGRP:

Calcitonin gene-related peptide

IR:

Insulin resistance

IDE:

Insulin-degrading enzyme

NEP:

Neprilysin

RAMPs:

Receptor activity modifying proteins

ROS:

Reactive oxygen species

Hsp70:

Heat shock protein 70

ER:

Endoplasmic reticulum

HSPG:

Heparan sulfate proteoglycan

NF-κB:

Nuclear factor kappa B

NFTs:

Neurofibrillary tangles

BBB:

Blood-brain barrier

CSF:

Cerebrospinal fluid

NLRP3:

Node like receptor pyrin 3

RAMP3:

Receptor activity modifying protein 3

APP:

Amyloid precursor protein

T3DM:

Type 3 diabetes mellitus

AMPK:

Adenosine monophosphate kinase protein kinase

IRS-1:

Insulin receptor substrate 1

GSK3β:

Glycogen synthase kinase 3 β

AGEs:

Advanced glycation end-products

TZDs:

Thiazolidinediones

PPARγ:

Peroxisome proliferator–activated receptor γ

DPP4:

Dipeptidyl peptidase 4

GLP-1:

Glucagon-like peptide-1

PI3K:

Phosphoinositide-3 kinase

References

  1. Bishoyi AK, Roham PH, Rachineni K, Save S, Hazari MA, Sharma S, et al. Human islet amyloid polypeptide (hIAPP)-a curse in type II diabetes mellitus: insights from structure and toxicity studies. Biol Chem. 2021;402(2):133–53.

    Article  CAS  PubMed  Google Scholar 

  2. El Saghir A, Farrugia G, Vassallo N. The human islet amyloid polypeptide in protein misfolding disorders: mechanisms of aggregation and interaction with biomembranes. Chem Phys Lipids. 2021;234:105010.

    Article  PubMed  Google Scholar 

  3. Tang Y, Zhang D, Zhang Y, Liu Y, Gong X, Chang Y, et al. Introduction and fundamentals of human islet amyloid polypeptide inhibitors. ACS Appl Bio Mater. 2020;3(12):8286–308.

    Article  CAS  PubMed  Google Scholar 

  4. Westermark P, Andersson A, Westermark GT. Islet amyloid polypeptide, islet amyloid, and diabetes mellitus. Physiol Rev. 2011;91(3):795–826.

    Article  CAS  PubMed  Google Scholar 

  5. Ling W, Huang Y-M, Qiao Y-C, Zhang X-X, Zhao H-L. Human amylin: from pathology to physiology and pharmacology. Curr Protein Pept Sci. 2019;20(9):944–57.

    Article  CAS  PubMed  Google Scholar 

  6. Cohen AS, Calkins E. Electron microscopic observations on a fibrous component in amyloid of diverse origins. Nature. 1959;183(4669):1202–3.

    Article  CAS  PubMed  Google Scholar 

  7. Pearse A, Ewen S, Polak J. The genesis of apudamyloid in endocrine polypeptide tumours: histochemical distinction from immunamyloid. Virchows Archiv B. 1972;10:93–107.

    CAS  Google Scholar 

  8. Westermark P, Wernstedt C, Wilander E, Sletten K. A novel peptide in the calcitonin gene related peptide family as an amyloid fibril protein in the endocrine pancreas. Biochem Biophys Res Commun. 1986;140(3):827–31.

    Article  CAS  PubMed  Google Scholar 

  9. Cooper G, Willis A, Clark A, Turner R, Sim R, Reid K. Purification and characterization of a peptide from amyloid-rich pancreases of type 2 diabetic patients. Proceedings of the National Academy of Sciences. 1987;84(23):8628-32.

  10. Cooper G, Leighton B, Dimitriadis G, Parry-Billings M, Kowalchuk J, Howland K et al. Amylin found in amyloid deposits in human type 2 diabetes mellitus may be a hormone that regulates glycogen metabolism in skeletal muscle. Proceedings of the National Academy of Sciences. 1988;85(20):7763-6.

  11. Hirai Y. In-vivo like studies of the h-IAPP amyloid precursors using Dielectric Relaxation. WORCESTER POLYTECHNIC INSTITUTE; 2013.

  12. Park YJ. The role of Fas-mediated apoptotic pathway in amyloid-induced beta-cell death. University of British Columbia; 2015.

  13. Zou X, Ouyang H, Yu T, Chen X, Pang D, Tang X, et al. Preparation of a new type 2 diabetic miniature pig model via the CRISPR/Cas9 system. Cell Death Dis. 2019;10(11):823.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Bhowmick DC. The molecular mechanisms of Stress-Induced Amylin turnover in pancreatic Beta cell. The George Washington University; 2018.

  15. Gilead S, Wolfenson H, Gazit E. Molecular mapping of the recognition interface between the islet amyloid polypeptide and insulin. Angew Chem. 2006;118(39):6626–30.

    Article  Google Scholar 

  16. Dubey R, Patil K, Dantu SC, Sardesai DM, Bhatia P, Malik N, et al. Azadirachtin inhibits amyloid formation, disaggregates pre-formed fibrils and protects pancreatic β-cells from human islet amyloid polypeptide/amylin-induced cytotoxicity. Biochem J. 2019;476(5):889–907.

    Article  CAS  PubMed  Google Scholar 

  17. Riahi Y, Wikstrom JD, Bachar-Wikstrom E, Polin N, Zucker H, Lee M-S, et al. Autophagy is a major regulator of beta cell insulin homeostasis. Diabetologia. 2016;59:1480–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Leissring MA, González-Casimiro CM, Merino B, Suire CN, Perdomo G. Targeting insulin-degrading enzyme in insulin clearance. Int J Mol Sci. 2021;22(5):2235.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. AlAnazi FH, Al-kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Ogaly HA et al. Effects of neprilysin and neprilysin inhibitors on glucose homeostasis: Controversial points and a promising arena. J Diabetes.n/a(n/a).

  20. Guillemain G, Lacapere J-J, Khemtemourian L. Targeting hIAPP fibrillation: A new paradigm to prevent β-cell death? Biochimica et Biophysica Acta (BBA)-Biomembranes. 2022:184002.

  21. Sevcuka A, White K, Terry C. Factors that contribute to hiapp amyloidosis in type 2 diabetes mellitus. Life. 2022;12(4):583.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rasheed HA, Al-Kuraishy HM, Al-Gareeb AI, Hussien NR, Al-Nami MS. Effects of diabetic pharmacotherapy on prolactin hormone in patients with type 2 diabetes mellitus: bane or Boon. J Adv Pharm Technol Res. 2019;10(4):163.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Al-Kuraishy HM, Al-Gareeb AI, Waheed HJ, Al-Maiahy TJ. Differential effect of metformin and/or glyburide on apelin serum levels in patients with type 2 diabetes mellitus: concepts and clinical practice. J Adv Pharm Technol Res. 2018;9(3):80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Al-kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Nadwa EH, Albogami SM et al. Metformin and growth differentiation factor 15 (GDF15) in type 2 diabetes mellitus: A hidden treasure. J Diabetes. 2022; n/a(n/a)

  25. Abdul-Hadi MH, Naji MT, Shams HA, Sami OM, Al-Harchan NA-A, Al-Kuraishy HM, et al. Oxidative stress injury and glucolipotoxicity in type 2 diabetes mellitus: the potential role of metformin and sitagliptin. Biomedical and Biotechnology Research Journal (BBRJ). 2020;4(2):166.

    Google Scholar 

  26. Al-Nami MS, Al-Kuraishy HM, Al-Gareeb AI, Al-Mamoori F. Metabolic profile and prolactin serum levels in men with type 2 diabetes mellitus: old-new rubric. Int J Crit Illn Inj Sci. 2019;9(3):120.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Mulder H, Gebre-Medhin S, Betsholtz C, Sundler F, Ahrén B. Islet amyloid polypeptide (amylin)-deficient mice develop a more severe form of alloxan-induced diabetes. Am J Physiology-Endocrinology Metabolism. 2000;278(4):E684–E91.

    Article  CAS  Google Scholar 

  28. Sanke T, Hanabusa T, Nakano Y, Oki C, Okai K, Nishimura S, et al. Plasma islet amyloid polypeptide (Amylin) levels and their responses to oral glucose in type 2 (non-insulin-dependent) diabetic patients. Diabetologia. 1991;34:129–32.

    Article  CAS  PubMed  Google Scholar 

  29. Bennett RG, Duckworth WC, Hamel FG. Degradation of amylin by insulin-degrading enzyme. J Biol Chem. 2000;275(47):36621–5.

    Article  CAS  PubMed  Google Scholar 

  30. Bennett RG, Hamel FG, Duckworth WC. An insulin-degrading enzyme inhibitor decreases amylin degradation, increases amylin-induced cytotoxicity, and increases amyloid formation in insulinoma cell cultures. Diabetes. 2003;52(9):2315–20.

    Article  CAS  PubMed  Google Scholar 

  31. Maianti JP, McFedries A, Foda ZH, Kleiner RE, Du XQ, Leissring MA, et al. Anti-diabetic activity of insulin-degrading enzyme inhibitors mediated by multiple hormones. Nature. 2014;511(7507):94–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tang W-J. Targeting insulin-degrading enzyme to treat type 2 diabetes mellitus. Trends Endocrinol Metab. 2016;27(1):24–34.

    Article  CAS  PubMed  Google Scholar 

  33. Bortoletto AS, Graham WV, Trout G, Bonito-Oliva A, Kazmi MA, Gong J et al. Human Islet Amyloid Polypeptide (hIAPP) Protofibril‐Specific Antibodies for Detection and Treatment of Type 2 Diabetes. Adv Sci. 2022:2202342.

  34. Clark A, Nilsson M. Islet amyloid: a complication of islet dysfunction or an aetiological factor in type 2 diabetes? Diabetologia. 2004;47:157–69.

    Article  CAS  PubMed  Google Scholar 

  35. Kahn SE, Andrikopoulos S, Verchere CB. Islet amyloid: a long-recognized but underappreciated pathological feature of type 2 diabetes. Diabetes. 1999;48(2):241–53.

    Article  CAS  PubMed  Google Scholar 

  36. Westermark P. Amyloid and polypeptide hormones: what is their interrelationship? Amyloid. 1994;1(1):47–60.

    Article  CAS  Google Scholar 

  37. Bram Y, Frydman-Marom A, Yanai I, Gilead S, Shaltiel-Karyo R, Amdursky N, et al. Apoptosis induced by islet amyloid polypeptide soluble oligomers is neutralized by diabetes-associated specific antibodies. Sci Rep. 2014;4(1):4267.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ma L, Yang C, Huang L, Chen Y, Li Y, Cheng C, et al. Glycated insulin exacerbates the cytotoxicity of human islet amyloid polypeptides: a vicious cycle in type 2 diabetes. ACS Chem Biol. 2019;14(3):486–96.

    Article  CAS  PubMed  Google Scholar 

  39. Hoenig M, Hall G, Ferguson D, Jordan K, Henson M, Johnson K, et al. A feline model of experimentally induced islet amyloidosis. Am J Pathol. 2000;157(6):2143–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Soty M, Visa M, Soriano S, del Carmen Carmona M, Nadal Á, Novials A. Involvement of ATP-sensitive potassium (KATP) channels in the loss of beta-cell function induced by human islet amyloid polypeptide. J Biol Chem. 2011;286(47):40857–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hussien NR, Al-Naimi MS, Rasheed HA, Al-Kuraishy HM, Al-Gareeb AI. Sulfonylurea and neuroprotection: the bright side of the moon. J Adv Pharm Technol Res. 2018;9(4):120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Xu J, Jin L, Chen J, Zhang R, Zhang H, Li Y, et al. Common variants in genes involved in islet amyloid polypeptide (IAPP) processing and the degradation pathway are associated with T2DM risk: a chinese population study. Diabetes Res Clin Pract. 2022;185:109235.

    Article  CAS  PubMed  Google Scholar 

  43. Jeong HR, An SSA. Causative factors for formation of toxic islet amyloid polypeptide oligomer in type 2 diabetes mellitus. Clin Interv Aging. 2015:1873–9.

  44. Fernández MS. Human IAPP amyloidogenic properties and pancreatic β-cell death. Cell Calcium. 2014;56(5):416–27.

    Article  PubMed  Google Scholar 

  45. Dubey R, Kulkarni SH, Dantu SC, Panigrahi R, Sardesai DM, Malik N, et al. Myricetin protects pancreatic β-cells from human islet amyloid polypeptide (hIAPP) induced cytotoxicity and restores islet function. Biol Chem. 2021;402(2):179–94.

    Article  CAS  PubMed  Google Scholar 

  46. Gurlo T, Ryazantsev S, Huang C-j, Yeh MW, Reber HA, Hines OJ, et al. Evidence for proteotoxicity in β cells in type 2 diabetes: toxic islet amyloid polypeptide oligomers form intracellularly in the secretory pathway. Am J Pathol. 2010;176(2):861–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sakagashira S, Hiddinga HJ, Tateishi K, Sanke T, Hanabusa T, Nanjo K, et al. S20G mutant amylin exhibits increased in vitro amyloidogenicity and increased intracellular cytotoxicity compared to wild-type amylin. Am J Pathol. 2000;157(6):2101–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Marek P, Abedini A, Song B, Kanungo M, Johnson ME, Gupta R, et al. Aromatic interactions are not required for amyloid fibril formation by islet amyloid polypeptide but do influence the rate of fibril formation and fibril morphology. Biochemistry. 2007;46(11):3255–61.

    Article  CAS  PubMed  Google Scholar 

  49. Wang J, Xu J, Finnerty J, Furuta M, Steiner DF, Verchere CB. The prohormone convertase enzyme 2 (PC2) is essential for processing pro-islet amyloid polypeptide at the NH2-terminal cleavage site. Diabetes. 2001;50(3):534–9.

    Article  CAS  PubMed  Google Scholar 

  50. Nilsson MR, Driscoll M, Raleigh DP. Low levels of asparagine deamidation can have a dramatic effect on aggregation of amyloidogenic peptides: implications for the study of amyloid formation. Protein Sci. 2002;11(2):342–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Monsellier E, Ramazzotti M, Taddei N, Chiti F. Aggregation propensity of the human proteome. PLoS Comput Biol. 2008;4(10):e1000199.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Tanaka A, Kaneto H, Miyatsuka T, Yamamoto K, Yoshiuchi K, Yamasaki Y, et al. Role of copper ion in the pathogenesis of type 2 diabetes. Endocr J. 2009;56(5):699–706.

    Article  CAS  PubMed  Google Scholar 

  53. Alexandrescu AT, Jha S, Patil SM, Gibson J, Alder NN, Nelson CE. Effects of heparin on amylin fibrillization. Biophys J. 2012;102(3):243a.

    Article  Google Scholar 

  54. Pilkington EH, Gurzov EN, Kakinen A, Litwak SA, Stanley WJ, Davis TP, et al. Pancreatic β-cell membrane fluidity and toxicity induced by human islet amyloid polypeptide species. Sci Rep. 2016;6(1):1–10.

    Article  Google Scholar 

  55. Brender JR, Krishnamoorthy J, Sciacca MF, Vivekanandan S, D’Urso L, Chen J, et al. Probing the sources of the apparent irreproducibility of amyloid formation: drastic changes in kinetics and a switch in mechanism due to micellelike oligomer formation at critical concentrations of IAPP. J Phys Chem B. 2015;119(7):2886–96.

    Article  CAS  PubMed  Google Scholar 

  56. Casas S, Gomis R, Gribble FM, Altirriba J, Knuutila S, Novials A. Impairment of the ubiquitin-proteasome pathway is a downstream endoplasmic reticulum stress response induced by extracellular human islet amyloid polypeptide and contributes to pancreatic β-cell apoptosis. Diabetes. 2007;56(9):2284–94.

    Article  CAS  PubMed  Google Scholar 

  57. Gurlo T, Costes S, Hoang JD, Rivera JF, Butler AE, Butler PC. β Cell–specific increased expression of calpastatin prevents diabetes induced by islet amyloid polypeptide toxicity. JCI insight. 2016;1(18).

  58. Milardi D, Gazit E, Radford SE, Xu Y, Gallardo RU, Caflisch A, et al. Proteostasis of islet amyloid polypeptide: a molecular perspective of risk factors and protective strategies for type II diabetes. Chem Rev. 2021;121(3):1845–93.

    Article  CAS  PubMed  Google Scholar 

  59. Nakhjavani M, Morteza A, Khajeali L, Esteghamati A, Khalilzadeh O, Asgarani F, et al. Increased serum HSP70 levels are associated with the duration of diabetes. Cell Stress Chaperones. 2010;15:959–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ladjimi M, Chaari A, Eliezer D. Inhibition of Human Islet Amyloid Polypeptide Aggregation in Type 2 Diabetes by Hsp70 Molecular Chaperones. Qatar Foundation Annual Research Conference Proceedings; 2016: HBKU Press Qatar; 2016. p. HBPP2702.

  61. Templin AT, Mellati M, Soininen R, Hogan MF, Esser N, Castillo JJ, et al. Loss of perlecan heparan sulfate glycosaminoglycans lowers body weight and decreases islet amyloid deposition in human islet amyloid polypeptide transgenic mice. Protein Eng Des Selection. 2019;32(2):95–102.

    Article  CAS  Google Scholar 

  62. Javed I, He J, Kakinen A, Faridi A, Yang W, Davis TP, et al. Probing the aggregation and immune response of human islet amyloid polypeptides with ligand-stabilized gold nanoparticles. ACS Appl Mater Interfaces. 2019;11(11):10462–71.

    Article  CAS  PubMed  Google Scholar 

  63. Westwell-Roper C, Denroche HC, Ehses JA, Verchere CB. Differential activation of innate immune pathways by distinct islet amyloid polypeptide (IAPP) aggregates. J Biol Chem. 2016;291(17):8908–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Westwell-Roper C, Dai DL, Soukhatcheva G, Potter KJ, van Rooijen N, Ehses JA, et al. IL-1 blockade attenuates islet amyloid polypeptide-induced proinflammatory cytokine release and pancreatic islet graft dysfunction. J Immunol. 2011;187(5):2755–65.

    Article  CAS  PubMed  Google Scholar 

  65. Armiento V, Hille K, Naltsas D, Lin JS, Barron AE, Kapurniotu A. The human host-defense peptide cathelicidin LL‐37 is a nanomolar inhibitor of amyloid self‐assembly of islet amyloid polypeptide (IAPP). Angew Chem Int Ed. 2020;59(31):12837–41.

    Article  CAS  Google Scholar 

  66. Salamekh S, Brender JR, Hyung S-J, Nanga RPR, Vivekanandan S, Ruotolo BT, et al. A two-site mechanism for the inhibition of IAPP amyloidogenesis by zinc. J Mol Biol. 2011;410(2):294–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Khemtemourian L, Antoniciello F, Sahoo BR, Decossas M, Lecomte S, Ramamoorthy A. Investigation of the effects of two major secretory granules components, insulin and zinc, on human-IAPP amyloid aggregation and membrane damage. Chem Phys Lipids. 2021;237:105083.

    Article  CAS  PubMed  Google Scholar 

  68. Brender JR, Hartman K, Nanga RPR, Popovych N, de la Salud Bea R, Vivekanandan S, et al. Role of zinc in human islet amyloid polypeptide aggregation. J Am Chem Soc. 2010;132(26):8973–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Farooq DM, Alamri AF, Alwhahabi BK, Metwally AM, Kareem KA. The status of zinc in type 2 diabetic patients and its association with glycemic control. J Family Community Med. 2020;27(1):29.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Alsubaie N, Al-Kuraishy HM, Al-Gareeb AI, Alharbi B, De Waard M, Sabatier J-M, et al. Statins use in alzheimer disease: bane or boon from frantic search and narrative review. Brain Sci. 2022;12(10):1290.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Al-kuraishy HM, Al-Gareeb AI, Saad HM, Batiha GE-S. Benzodiazepines in Alzheimer’s disease: beneficial or detrimental effects. Inflammopharmacology. 2022.

  72. Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Hakami ZH, Khamjan NA, Saad HM et al. A Potential Link Between Visceral Obesity and Risk of Alzheimer’s Disease. Neurochem Res. 2022.

  73. Karvani M, Simos P, Stavrakaki S, Kapoukranidou D. Neurocognitive impairment in type 2 diabetes mellitus. Hormones. 2019;18:523–34.

    Article  PubMed  Google Scholar 

  74. Akomolafe A, Beiser A, Meigs JB, Au R, Green RC, Farrer LA, et al. Diabetes mellitus and risk of developing Alzheimer disease: results from the Framingham Study. Arch Neurol. 2006;63(11):1551–5.

    Article  PubMed  Google Scholar 

  75. Biessels GJ, Staekenborg S, Brunner E, Brayne C, Scheltens P. Risk of dementia in diabetes mellitus: a systematic review. Lancet Neurol. 2006;5(1):64–74.

    Article  PubMed  Google Scholar 

  76. Allen KV, Frier BM, Strachan MW. The relationship between type 2 diabetes and cognitive dysfunction: longitudinal studies and their methodological limitations. Eur J Pharmacol. 2004;490(1–3):169–75.

    Article  CAS  PubMed  Google Scholar 

  77. Arvanitakis Z, Wilson RS, Bienias JL, Evans DA, Bennett DA. Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. Arch Neurol. 2004;61(5):661–6.

    Article  PubMed  Google Scholar 

  78. Janson J, Laedtke T, Parisi JE, O’Brien P, Petersen RC, Butler PC. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes. 2004;53(2):474–81.

    Article  CAS  PubMed  Google Scholar 

  79. De Felice FG, Gonçalves RA, Ferreira ST. Impaired insulin signalling and allostatic load in Alzheimer disease. Nat Rev Neurosci. 2022;23(4):215–30.

    Article  PubMed  Google Scholar 

  80. Teixeira MM, de Azeredo Passos VM, Barreto SM, Schmidt MI, Duncan BB, Beleigoli AM, et al. Markers of adiposity, insulin resistance, prediabetes and cognitive function at baseline of the brazilian longitudinal study of Adult Health (ELSA–Brasil). Diabetes Res Clin Pract. 2020;170:108499.

    Article  Google Scholar 

  81. Ma L, Yang C, Zheng J, Chen Y, Xiao Y, Huang K. Non-polyphenolic natural inhibitors of amyloid aggregation. Eur J Med Chem. 2020;192:112197.

    Article  CAS  PubMed  Google Scholar 

  82. Zhang Y, Song W. Islet amyloid polypeptide: another key molecule in Alzheimer’s pathogenesis? Prog Neurobiol. 2017;153:100–20.

    Article  CAS  PubMed  Google Scholar 

  83. Fawver N, Ghiwot J, Koola Y, Carrera C, Rodriguez-Rivera W, Hernandez J. Islet amyloid polypeptide (IAPP): a second amyloid in Alzheimer’s disease. Curr Alzheimer Res. 2014;11(10):928–40.

    Article  CAS  PubMed  Google Scholar 

  84. Krotee P, Griner SL, Sawaya MR, Cascio D, Rodriguez JA, Shi D, et al. Common fibrillar spines of amyloid-β and human islet amyloid polypeptide revealed by microelectron diffraction and structure-based inhibitors. J Biol Chem. 2018;293(8):2888–902.

    Article  CAS  PubMed  Google Scholar 

  85. Hu R, Zhang M, Chen H, Jiang B, Zheng J. Cross-seeding interaction between β-amyloid and human islet amyloid polypeptide. ACS Chem Neurosci. 2015;6(10):1759–68.

    Article  CAS  PubMed  Google Scholar 

  86. Jackson K, Barisone GA, Diaz E, Jin Lw, DeCarli C, Despa F. Amylin deposition in the brain: a second amyloid in Alzheimer disease? Ann Neurol. 2013;74(4):517–26.

    Article  CAS  PubMed  Google Scholar 

  87. Oskarsson ME, Paulsson JF, Schultz SW, Ingelsson M, Westermark P, Westermark GT. In vivo seeding and cross-seeding of localized amyloidosis: a molecular link between type 2 diabetes and Alzheimer disease. Am J Pathol. 2015;185(3):834–46.

    Article  CAS  PubMed  Google Scholar 

  88. Ly H, Verma N, Sharma S, Kotiya D, Despa S, Abner EL et al. The association of circulating amylin with β-amyloid in familial Alzheimer’s disease. Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2021;7(1):e12130.

  89. Miklossy J, Qing H, Radenovic A, Kis A, Vileno B, Làszló F, et al. Beta amyloid and hyperphosphorylated tau deposits in the pancreas in type 2 diabetes. Neurobiol Aging. 2010;31(9):1503–15.

    Article  CAS  PubMed  Google Scholar 

  90. Schultz N, Janelidze S, Byman E, Minthon L, Nägga K, Hansson O, et al. Levels of islet amyloid polypeptide in cerebrospinal fluid and plasma from patients with Alzheimer’s disease. PLoS ONE. 2019;14(6):e0218561.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Götz J, Lim Y-A, Eckert A. Lessons from two prevalent amyloidoses—what amylin and Aβ have in common. Front Aging Neurosci. 2013;5:38.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Ferreira S, Raimundo AF, Menezes R, Martins IC. Islet amyloid polypeptide & amyloid beta peptide roles in Alzheimer’s disease: two triggers, one disease. Neural regeneration research. 2021;16(6):1127.

    Article  CAS  PubMed  Google Scholar 

  93. Raimundo AF, Ferreira S, Martins IC, Menezes R. Islet amyloid polypeptide: A Partner in Crime with Aβ in the Pathology of Alzheimer’s Disease. Front Mol Neurosci. 2020;13:35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wijesekara N, Ahrens R, Sabale M, Wu L, Ha K, Verdile G, et al. Amyloid-β and islet amyloid pathologies link Alzheimer’s disease and type 2 diabetes in a transgenic model. FASEB J. 2017;31(12):5409–18.

    Article  CAS  PubMed  Google Scholar 

  95. Zhang G, Meng L, Wang Z, Peng Q, Chen G, Xiong J, et al. Islet amyloid polypeptide cross-seeds tau and drives the neurofibrillary pathology in Alzheimer’s disease. Mol Neurodegener. 2022;17(1):12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Murray ME, Kouri N, Lin W-L, Jack CR, Dickson DW, Vemuri P. Clinicopathologic assessment and imaging of tauopathies in neurodegenerative dementias. Alzheimers Res Ther. 2014;6(1):1–13.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Patrick S, Corrigan R, Grizzanti J, Mey M, Blair J, Pallas M, et al. Neuroprotective effects of the amylin analog, pramlintide, on Alzheimer’s disease are associated with oxidative stress regulation mechanisms. J Alzheimers Dis. 2019;69(1):157–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Fox A, Snollaerts T, Errecart Casanova C, Calciano A, Nogaj LA, Moffet DA. Selection for nonamyloidogenic mutants of islet amyloid polypeptide (IAPP) identifies an extended region for amyloidogenicity. Biochemistry. 2010;49(36):7783–9.

    Article  CAS  PubMed  Google Scholar 

  99. García-Viñuales S, Sciacca MF, Lanza V, Santoro AM, Grasso G, Tundo GR, et al. The interplay between lipid and Aβ amyloid homeostasis in Alzheimer’s Disease: risk factors and therapeutic opportunities. Chem Phys Lipids. 2021;236:105072.

    Article  PubMed  Google Scholar 

  100. Yang Y-Y, Ren Y-T, Jia M-Y, Bai C-Y, Liang X-T, Gao H-L, et al. The human islet amyloid polypeptide reduces hippocampal tauopathy and behavioral impairments in P301S mice without inducing neurotoxicity or seeding amyloid aggregation. Exp Neurol. 2023;362:114346.

    Article  CAS  PubMed  Google Scholar 

  101. Thota RN, Rosato JI, Dias CB, Burrows TL, Martins RN, Garg ML. Dietary supplementation with curcumin reduce circulating levels of glycogen synthase kinase-3β and islet amyloid polypeptide in adults with high risk of type 2 diabetes and Alzheimer’s disease. Nutrients. 2020;12(4):1032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Bharadwaj P, Solomon T, Sahoo BR, Ignasiak K, Gaskin S, Rowles J, et al. Amylin and beta amyloid proteins interact to form amorphous heterocomplexes with enhanced toxicity in neuronal cells. Sci Rep. 2020;10(1):1–14.

    Article  Google Scholar 

  103. Jhamandas JH, Li Z, Westaway D, Yang J, Jassar S, MacTavish D. Actions of β-amyloid protein on human neurons are expressed through the amylin receptor. Am J Pathol. 2011;178(1):140–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wright S, Malinin NL, Powell KA, Yednock T, Rydel RE, Griswold-Prenner I. α2β1 and αVβ1 integrin signaling pathways mediate amyloid-β-induced neurotoxicity. Neurobiol Aging. 2007;28(2):226–37.

    Article  CAS  PubMed  Google Scholar 

  105. Sciacca MF, Tempra C, Scollo F, Milardi D, La Rosa C. Amyloid growth and membrane damage: Current themes and emerging perspectives from theory and experiments on Aβ and hIAPP. Biochimica et Biophysica Acta (BBA)-Biomembranes. 2018;1860(9):1625–38.

  106. Nakamura T, Oh C-k, Zhang X, Lipton SA. Protein S-nitrosylation and oxidation contribute to protein misfolding in neurodegeneration. Free Radic Biol Med. 2021;172:562–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Wang X, Wang W, Li L, Perry G, Lee H-g, Zhu X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim et Biophys Acta (BBA)-Molecular Basis Disease. 2014;1842(8):1240–7.

    Article  CAS  Google Scholar 

  108. Roham PH, Save SN, Sharma S. Human islet amyloid polypeptide: A therapeutic target for the management of type 2 diabetes mellitus. J Pharm Anal. 2022.

  109. Xu X, Sun Y, Cen X, Shan B, Zhao Q, Xie T, et al. Metformin activates chaperone-mediated autophagy and improves disease pathologies in an Alzheimer disease mouse model. Protein Cell. 2021;12(10):769–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Borchi E, Bargelli V, Guidotti V, Berti A, Stefani M, Nediani C, et al. Mild exposure of RIN-5F β-cells to human islet amyloid polypeptide aggregates upregulates antioxidant enzymes via NADPH oxidase-RAGE: an hormetic stimulus. Redox Biol. 2014;2:114–22.

    Article  CAS  Google Scholar 

  111. Conde-Knape K. Heparan sulfate proteoglycans in experimental models of diabetes: a role for perlecan in diabetes complications. Diabetes Metab Res Rev. 2001;17(6):412–21.

    Article  CAS  PubMed  Google Scholar 

  112. Hiebert LM. Heparan sulfate proteoglycans in diabetes. Semin Thromb Hemost; 2021: Thieme Medical Publishers, Inc.; 2021. p. 261 – 73.

  113. Van Horssen J, Wesseling P, Van Den Heuvel LP, De Waal RM, Verbeek MM. Heparan sulphate proteoglycans in Alzheimer’s disease and amyloid-related disorders. Lancet Neurol. 2003;2(8):482–92.

    Article  PubMed  Google Scholar 

  114. Lorente-Gea L, García B, Martín C, Ordiales H, García-Suárez O, Piña-Batista KM, et al. Heparan sulfate proteoglycans undergo differential expression alterations in Alzheimer disease brains. J Neuropathol Exp Neurol. 2020;79(5):474–83.

    Article  CAS  PubMed  Google Scholar 

  115. Tang Y, Zhang D, Gong X, Zheng J. A mechanistic survey of Alzheimer’s disease. Biophys Chem. 2022;281:106735.

    Article  CAS  PubMed  Google Scholar 

  116. Stamouli E, Politis A. Pro-inflammatory cytokines in Alzheimer’s disease. Psychiatrike = Psychiatriki. 2016;27(4):264–75.

    Article  CAS  PubMed  Google Scholar 

  117. Uddin MS, Hasana S, Ahmad J, Hossain M, Rahman M, Behl T, et al. Anti-neuroinflammatory potential of polyphenols by inhibiting NF-κB to halt Alzheimer’s disease. Curr Pharm Des. 2021;27(3):402–14.

    Article  CAS  PubMed  Google Scholar 

  118. Templin AT, Mellati M, Meier DT, Esser N, Hogan MF, Castillo JJ, et al. Low concentration IL-1β promotes islet amyloid formation by increasing hIAPP release from humanised mouse islets in vitro. Diabetologia. 2020;63:2385–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Xie Z, Wu H, Zhao J. Multifunctional roles of zinc in Alzheimer’s disease. Neurotoxicology. 2020;80:112–23.

    Article  CAS  PubMed  Google Scholar 

  120. Morris JK, Vidoni ED, Mahnken JD, Montgomery RN, Johnson DK, Thyfault JP, et al. Cognitively impaired elderly exhibit insulin resistance and no memory improvement with infused insulin. Neurobiol Aging. 2016;39:19–24.

    Article  CAS  PubMed  Google Scholar 

  121. Al-Kuraishy HM, Sami OM, Hussain NR, Al-Gareeb AI. Metformin and/or vildagliptin mitigate type II diabetes mellitus induced-oxidative stress: the intriguing effect. J Adv Pharm Technol Res. 2020;11(3):142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Inoue Y, Masuda T, Misumi Y, Ando Y, Ueda M. Metformin attenuates vascular pathology by increasing expression of insulin-degrading enzyme in a mixed model of cerebral amyloid angiopathy and type 2 diabetes mellitus. Neurosci Lett. 2021;762:136136.

    Article  CAS  PubMed  Google Scholar 

  123. Poor SR, Ettcheto M, Cano A, Sanchez-Lopez E, Manzine PR, Olloquequi J, et al. Metformin a potential pharmacological strategy in late onset Alzheimer’s disease treatment. Pharmaceuticals. 2021;14(9):890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Farr SA, Roesler E, Niehoff ML, Roby DA, McKee A, Morley JE. Metformin improves learning and memory in the SAMP8 mouse model of Alzheimer’s disease. J Alzheimers Dis. 2019;68(4):1699–710.

    Article  CAS  PubMed  Google Scholar 

  125. Koenig AM, Mechanic-Hamilton D, Xie SX, Combs MF, Cappola AR, Xie L, et al. Effects of the insulin sensitizer metformin in Alzheimer’s disease: pilot data from a randomized placebo-controlled crossover study. Alzheimer Dis Assoc Disord. 2017;31(2):107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Sluggett JK, Koponen M, Bell JS, Taipale H, Tanskanen A, Tiihonen J, et al. Metformin and risk of Alzheimer’s disease among community-dwelling people with diabetes: a national case-control study. J Clin Endocrinol Metabolism. 2020;105(4):e963–e72.

    Article  Google Scholar 

  127. Hsu C-C, Wahlqvist ML, Lee M-S, Tsai H-N. Incidence of dementia is increased in type 2 diabetes and reduced by the use of sulfonylureas and metformin. J Alzheimers Dis. 2011;24(3):485–93.

    Article  CAS  PubMed  Google Scholar 

  128. Ning P, Luo A, Mu X, Xu Y, Li T. Exploring the dual character of metformin in Alzheimer’s disease. Neuropharmacology. 2022:108966.

  129. Boura-Halfon S, Zick Y. Phosphorylation of IRS proteins, insulin action, and insulin resistance. Am J Physiology-Endocrinology Metabolism. 2009;296(4):E581–E91.

    Article  CAS  Google Scholar 

  130. Salminen A, Kaarniranta K, Haapasalo A, Soininen H, Hiltunen M. AMP-activated protein kinase: a potential player in Alzheimer’s disease. J Neurochem. 2011;118(4):460–74.

    Article  CAS  PubMed  Google Scholar 

  131. Luo A, Ning P, Lu H, Huang H, Shen Q, Zhang D et al. Association Between Metformin and Alzheimer’s Disease: A Systematic Review and Meta-Analysis of Clinical Observational Studies. J Alzheimers Dis. 2022(Preprint):1–13.

  132. Rodriguez-Rodriguez P, Sandebring-Matton A, Merino-Serrais P, Parrado-Fernandez C, Rabano A, Winblad B, et al. Tau hyperphosphorylation induces oligomeric insulin accumulation and insulin resistance in neurons. Brain. 2017;140(12):3269–85.

    Article  PubMed  Google Scholar 

  133. de la Monte SM, Tong M, Daiello LA, Ott BR. Early-stage Alzheimer’s disease is associated with simultaneous systemic and central nervous system dysregulation of insulin-linked metabolic pathways. J Alzheimers Dis. 2019;68(2):657–68.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Marciniak E, Leboucher A, Caron E, Ahmed T, Tailleux A, Dumont J, et al. Tau deletion promotes brain insulin resistance. J Exp Med. 2017;214(8):2257–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Hayden MR. Type 2 diabetes mellitus increases the risk of late-onset Alzheimer’s disease: ultrastructural remodeling of the neurovascular unit and diabetic gliopathy. Brain Sci. 2019;9(10):262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Huang W-H, Chen W, Jiang L-y, Yang Y-X, Yao L-F, Li K-S. Influence of ADAM10 polymorphisms on plasma level of soluble receptor for advanced glycation end products and the association with Alzheimer’s disease risk. Front Genet. 2018;9:540.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Sanati M, Aminyavari S, Afshari AR, Sahebkar A. Mechanistic insight into the role of metformin in Alzheimer’s disease. Life Sci. 2022:120299.

  138. Khezri MR, Yousefi K, Mahboubi N, Hodaei D, Ghasemnejad-Berenji M. Metformin in Alzheimer’s disease: An overview of potential mechanisms, preclinical and clinical findings. Biochem Pharmacol. 2022:114945.

  139. Lebovitz HE. Thiazolidinediones: the forgotten diabetes medications. Curr Diab Rep. 2019;19:1–13.

    Article  Google Scholar 

  140. Cam ME, Hazar-Yavuz AN, Yildiz S, Ertas B, Adakul BA, Taskin T, et al. The methanolic extract of Thymus praecox subsp. skorpilii var. skorpilii restores glucose homeostasis, ameliorates insulin resistance and improves pancreatic β-cell function on streptozotocin/nicotinamide-induced type 2 diabetic rats. J Ethnopharmacol. 2019;231:29–38.

    Article  CAS  PubMed  Google Scholar 

  141. Bae J, Kim G, Lee Y-H, Lee B-W, Kang ES, Cha B-S. Differential effects of thiazolidinediones and dipeptidyl peptidase-4 inhibitors on insulin resistance and β-cell function in type 2 diabetes mellitus: a propensity score-matched analysis. Diabetes Ther. 2019;10:149–58.

    Article  CAS  PubMed  Google Scholar 

  142. Hull RL, Shen Z-P, Watts MR, Kodama K, Carr DB, Utzschneider KM, et al. Long-term treatment with rosiglitazone and metformin reduces the extent of, but does not prevent, islet amyloid deposition in mice expressing the gene for human islet amyloid polypeptide. Diabetes. 2005;54(7):2235–44.

    Article  CAS  PubMed  Google Scholar 

  143. Andrikopoulos S, Verchere CB, Terauchi Y, Kadowaki T, Kahn SE. beta-cell glucokinase deficiency and hyperglycemia are associated with reduced islet amyloid deposition in a mouse model of type 2 diabetes. Diabetes. 2000;49(12):2056–62.

    Article  CAS  PubMed  Google Scholar 

  144. Craig A, Parvez F, Issberner J. A systematic literature review of the effect of insulin sensitizers on the cognitive symptoms of Alzheimer’s Disease in transgenic mice. Behav Brain Res. 2019;372:112015.

    Article  CAS  PubMed  Google Scholar 

  145. Miller BW, Willett KC, Desilets AR. Rosiglitazone and pioglitazone for the treatment of Alzheimer’s disease. Ann Pharmacother. 2011;45(11):1416–24.

    Article  CAS  PubMed  Google Scholar 

  146. Watson GS, Cholerton BA, Reger MA, Baker LD, Plymate SR, Asthana S, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J geriatric psychiatry. 2005;13(11):950–8.

    Google Scholar 

  147. Gold M, Alderton C, Zvartau-Hind M, Egginton S, Saunders AM, Irizarry M, et al. Rosiglitazone monotherapy in mild-to-moderate Alzheimer’s disease: results from a randomized, double-blind, placebo-controlled phase III study. Dement Geriatr Cogn Disord. 2010;30(2):131–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Alomair BM, Al-Kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, De Waard M, Elekhnawy E et al. Is sitagliptin effective for SARS-CoV-2 infection: false or true prophecy? Inflammopharmacology. 2022:1–5.

  149. Al-Kuraishy HM, Al-Gareeb AI, Qusty N, Alexiou A, Batiha GE-S. Impact of Sitagliptin on non-diabetic Covid-19 patients. Curr Mol Pharmacol. 2022;15(4):683–92.

    Article  CAS  PubMed  Google Scholar 

  150. d’Amico M, Di Filippo C, Marfella R, Abbatecola AM, Ferraraccio F, Rossi F, et al. Long-term inhibition of dipeptidyl peptidase-4 in Alzheimer’s prone mice. Exp Gerontol. 2010;45(3):202–7.

    Article  PubMed  Google Scholar 

  151. Kosaraju J, Gali CC, Khatwal RB, Dubala A, Chinni S, Holsinger RD, et al. Saxagliptin: a dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer’s disease. Neuropharmacology. 2013;72:291–300.

    Article  CAS  PubMed  Google Scholar 

  152. Ikeda Y, Nagase N, Tsuji A, Kitagishi Y, Matsuda S. Neuroprotection by dipeptidyl-peptidase-4 inhibitors and glucagon-like peptide-1 analogs via the modulation of AKT-signaling pathway in Alzheimer’s disease. World J Biol Chem. 2021;12(6):104.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Solmaz V, Çınar BP, Yiğittürk G, Çavuşoğlu T, Taşkıran D, Erbaş O. Exenatide reduces TNF-α expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur J Pharmacol. 2015;765:482–7.

    Article  CAS  PubMed  Google Scholar 

  154. Cai H-Y, Yang J-T, Wang Z-J, Zhang J, Yang W, Wu M-N, et al. Lixisenatide reduces amyloid plaques, neurofibrillary tangles and neuroinflammation in an APP/PS1/tau mouse model of Alzheimer’s disease. Biochem Biophys Res Commun. 2018;495(1):1034–40.

    Article  CAS  PubMed  Google Scholar 

  155. Friedman LG, Qureshi YH, Yu WH. Promoting autophagic clearance: viable therapeutic targets in Alzheimer’s disease. Neurotherapeutics. 2015;12:94–108.

    Article  CAS  PubMed  Google Scholar 

  156. Ahrén B, Winzell MS, Wierup N, Sundler F, Burkey B, Hughes TE. DPP-4 inhibition improves glucose tolerance and increases insulin and GLP-1 responses to gastric glucose in association with normalized islet topography in mice with β-cell-specific overexpression of human islet amyloid polypeptide. Regul Pept. 2007;143(1–3):97–103.

    Article  PubMed  Google Scholar 

  157. Matveyenko AV, Dry S, Cox HI, Moshtaghian A, Gurlo T, Galasso R, et al. Beneficial endocrine but adverse exocrine effects of sitagliptin in the human islet amyloid polypeptide transgenic rat model of type 2 diabetes: interactions with metformin. Diabetes. 2009;58(7):1604–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Deanship of Scientific Research at Shaqra University for supporting this work.

Funding

Open Access funding enabled and organized by Projekt DEAL. This work was supported by the University of Witten-Herdecke Germany.

Open Access funding enabled and organized by Projekt DEAL.

Author information

Authors and Affiliations

Authors

Contributions

HMA and AIA: conceptualization, data collection, and writing of the manuscript. MA, AA, MP, H.M.S, and G.E.B: writing, supervision, figures preparation, reviewing, editing of the manuscript and responding to reviewers’ comments. All authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to Marios Papadakis.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alrouji, M., Al-Kuraishy, H.M., Al-Gareeb, A.I. et al. The potential role of human islet amyloid polypeptide in type 2 diabetes mellitus and Alzheimer’s diseases. Diabetol Metab Syndr 15, 101 (2023). https://doi.org/10.1186/s13098-023-01082-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13098-023-01082-1

Keywords