Next Article in Journal
Bromotyrosine-Derived Metabolites from a Marine Sponge Inhibit Pseudomonas aeruginosa Biofilms
Next Article in Special Issue
Exosomes; a Potential Source of Biomarkers, Therapy, and Cure for Type-1 Diabetes
Previous Article in Journal
The Lack of Synergy between Carvedilol and the Preventive Effect of Dexrazoxane in the Model of Chronic Anthracycline-Induced Cardiomyopathy
Previous Article in Special Issue
MSC−sEV Treatment Polarizes Pro−Fibrotic M2 Macrophages without Exacerbating Liver Fibrosis in NASH
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Pancreatic Tumour-Derived Exosomes and Their Cargo in Pancreatic Cancer-Related Diabetes

by
Helen B. Binang
1,2,
Chamini J. Perera
1,2,† and
Minoti V. Apte
1,2,*,†
1
Pancreatic Research Group, South Western Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
2
Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(12), 10203; https://doi.org/10.3390/ijms241210203
Submission received: 25 May 2023 / Revised: 10 June 2023 / Accepted: 13 June 2023 / Published: 15 June 2023
(This article belongs to the Special Issue Exosomes 2.0)

Abstract

:
One of the most common and deadly types of pancreatic cancer (PC) is pancreatic ductal adenocarcinoma (PDAC), with most patients succumbing to the disease within one year of diagnosis. Current detection strategies do not address asymptomatic PC; therefore, patients are diagnosed at an advanced stage when curative treatment is often no longer possible. In order to detect PC in asymptomatic patients earlier, the risk factors that could serve as reliable markers need to be examined. Diabetic mellitus (DM) is a significant risk factor for this malignancy and can be both a cause and consequence of PC. Typically, DM caused by PC is known as new-onset, pancreatogenic, pancreoprivic, or pancreatic cancer-related diabetes (PCRD). Although PCRD is quite distinct from type 2 DM (T2DM), there are currently no biomarkers that differentiate PCRD from T2DM. To identify such biomarkers, a better understanding of the mechanisms mediating PCRD is essential. To this end, there has been a growing research interest in recent years to elucidate the role of tumour-derived exosomes and their cargo in the pathogenesis of PCRD. Exosomes derived from tumours can be recognized for their specificity because they reflect the characteristics of their parent cells and are important in intercellular communication. Their cargo consists of proteins, lipids, and nucleic acids, which can be transferred to and alter the behaviour of recipient cells. This review provides a concise overview of current knowledge regarding tumour-derived exosomes and their cargo in PCRD and discusses the potential areas worthy of further study.

1. Introduction

Pancreatic cancer (PC) is the seventh leading cause of cancer-related deaths in the world, with the most common type, comprising >90% of PC cases, taking the form of pancreatic ductal adenocarcinoma (PDAC) [1,2,3]. The incidence of PC is around 45 out of every 100,000 people, and currently, the 5-year survival rate is dismal, hovering around 11.5% [4]. Deaths from PC are on the rise. For example, in Australia, PC was the fourth most common cause of cancer deaths in 2020 and the third most common cause of death from cancer in 2022 [4]. The disease is often silent in nature, and symptoms only manifest after it has progressed to an advanced stage. Indeed, the symptoms of PC are often non-specific, ranging from vague abdominal pain, general malaise, nausea and vomiting, poor appetite, and weight loss. The poor clinical outcomes in this disease are a result of several factors, including late diagnosis, early distant metastasis, resistance to most conventional treatment options, and a dense tumour microenvironment that inhibits the penetration of drugs into the tumour and actively facilitates tumour progression via a range of mechanisms [5].
The major risk factors for PC include older age, sex (high prevalence in men), smoking, a history of chronic pancreatitis (CP), gene mutations, obesity, and diabetes mellitus (DM) [6]. Obesity and CP are also associated with metabolic disorders such as insulin resistance and glucose intolerance, which are characteristics of diabetes [7,8]. However, as CP is a known cause of diabetes, whether the metabolic defects seen in obesity are causes or effects of diabetes and/or PC remains to be clearly established [9,10].

1.1. Pancreatic Cancer and Diabetes

Diabetes mellitus is a heterogeneous metabolic disorder that is characterized by the presence of hyperglycaemia due to the impairment of insulin secretion and/or defective insulin action leading to peripheral insulin resistance. It is a major public health problem that affects over 462 million people worldwide [11]. Its incidence is predicted to reach 7079 per 100,000 by 2030 and 7862 per 100,000 by 2040 [12]. It is associated with increased risks for stroke, blindness, amputations, heart disease, kidney disease and cancer. DM can be classified into three main categories, namely type 1 DM (T1DM), type 2 DM (T2DM), and gestational diabetes. A form of DM associated with diseases of the exocrine pancreas is known as type 3c DM (T3cDM) [13]. DM can be both a cause and a consequence of PC [14].

1.2. Pancreatic Cancer-Related Diabetes (PCRD)

A subset of PC patients reported being diagnosed with DM within 3–5 years before their diagnosis of PC. This recent onset of diabetes has also been termed pancreatic cancer-related diabetes (PCRD) and may be a harbinger of PC [9]. PCRD may be caused by mediators of β cell dysfunctions and peripheral insulin resistance, the interaction between PC and adipose tissue, the control of DM-associated PC pathway genes by miRNAs and the accelerated transforming growth factor β (TGF-β) signalling due to the increased secretion of TGF-β, causing the depletion of β cells [15,16,17,18,19,20,21,22,23,24,25,26,27,28]. There are distinct differences between the clinical picture of PCRD and that of type 2 DM (T2DM). Unlike T2DM, PCRD is characterized by weight loss and often normal insulin levels. Notably, the resolution of diabetes occurs after pancreatic tumour resection [29,30,31] (Table 1).
The underlying association between PC and DM is complicated due to the presence of a bidirectional link. The ability to distinguish DM caused by PC from T2DM is critical for recognizing that the new onset of DM may indicate a developing PC. Therefore, identifying novel, clinically applicable biomarkers of high sensitivity and specificity in the setting of PCRD may lead to earlier PC detection and subsequently improved therapeutic outcomes [32,33]. It is possible that developing cancer from its earliest stage, called pancreatic intraepithelial neoplasm (PanIN), secrete factors that are carried within exosomes (a type of extracellular vesicle, EV) to impair the function of islets and other peripheral cells, leading to decreased insulin secretion and/or peripheral insulin resistance. Thus, studies of tumour-derived exosomes and their cargo have recently become a focus of research in order to understand their role in PCRD.
Since the best-studied EVs in the literature are exosomes, this review focused on the possible role of exosomes as mediators of PCRD. We summarized the recent work on exosomes in relation to PC as well as DM and the specific roles of pancreatic tumour-derived exosomes in PCRD development. By unveiling the putative adverse influence of PC on systemic glucose metabolism resulting in PCRD, potential biomarkers could be uncovered to improve the early diagnosis and treatment of PC.

2. Exosomes

Exosomes are part of a family of membrane-derived particles that are collectively known as EVs. They facilitate intercellular communication (in health as well as in diseased states) via their cargo which consists of a mixture of bioactive molecules such as nucleic acids (mRNAs, miRNAs, DNA), lipids and proteins, which can be transported to the targeted cells/tissues via the blood or lymph circulation [34]. Based on their molecular sizes and mode of formation, EVs can be classified as apoptotic bodies, microvesicles and exosomes [35].

2.1. Exosome Formation and Characterization

Exosomes are small Evs (sEVs) that are contained within multivesicular bodies (MVBs) and are released when the MVBs fuse with the plasma membrane (Figure 1). Exosome biogenesis begins when the early-sorting endosome matures into a late-sorting endosome or MVB [36]. The MVBs contain intraluminal vesicles (ILVs), which are the exosome precursors. MVBs can either be degraded (via fusion with lysosomes) or transported to the plasma membrane for exosome release. The fusion of MVBs with the plasma membrane results in ILVs being released as exosomes into body fluids.
Exosomes are cup-shaped, cell-specific, small double-membraned EVs that are heterogeneous in size and range from 30 to 150 nm; they are produced by different cell types and can be found in all biological fluids. They are identified based on their size, density, morphology, and the presence of certain surface markers. The common surface markers of exosomes are tetraspanins (CD63, CD81 and CD9), fusion proteins (Flotilin, Annexins, GTPases), endosome-associated proteins (Alix, TSG101) and heat shock proteins (Hsp70, Hsp90) [37].

2.2. Uptake of Exosomes by Recipient Cells

Generally, cells may take up exosomes through a variety of endocytic pathways, namely: clathrin-independent pathways such as direct membrane fusion, macropinocytosis, phagocytosis or via clathrin-dependent endocytosis (Figure 2). Other pathways of exosome uptake include caveolin-mediated uptake and lipid raft-mediated internalization. The mechanism of this uptake adopted by target cells for a given exosome may depend on proteins and glycoproteins, which can be found on the surface of both the exosome and the target cell [38]. Upon being taken up by target cells, exosomes may influence cell function via the release of cargo molecules which can activate intracellular signalling. Exosomes and their cargo are involved in the pathogenic processes of both PC [39,40,41] and DM [42,43].
As noted earlier, the mechanisms of PCRD may be centred around the influence of pancreatic tumour-derived exosomes or the ability of β cells to produce insulin and that of other cells to produce, take up or utilize glucose.

3. Pancreatic Cancer-Derived Exosomes

The use of tumour-derived exosomes as liquid biopsies in cancer management is currently an attractive area of investigation with promising results [44]. This is because obtaining samples from patients for exosome isolation is minimally invasive as exosomes are present in body fluids such as blood, saliva, ascites or urine. In addition, exosomes are highly stable and specific, and the exosome cargo, namely, proteins, lipids and nucleic acids are highly reflective of parental cells. As such, it is possible to understand the behaviour of certain cells by examining the cargo contained in their exosomes. Exosomes produced under pathological conditions often exhibit tendencies that are different from those obtainable under normal conditions. In cancers, exosomes produced by tumour cells contain molecules that are cell-specific, and these molecules can be used to differentiate tumour cells from normal cells and hence, cancer patients from healthy individuals [45]. However, the processes surrounding these differences in the exosome composition among cells are not yet known, and the means by which these processes are regulated remains to be fully elucidated.

4. Role of Pancreatic Tumour-Derived Exosomes in PCRD

Exosomes originating from PC cells can transport cargo molecules to different cell types, ultimately affecting disease progression. PC-derived exosomes can regulate angiogenesis, deliver tissue factor (TF) promoting cancer-associated thrombosis, regulate immune functions, promote tumour growth, invasion and metastasis, confer chemoresistance, and regulate pancreatic functions. The role of tumour cell-derived exosomes in pancreatic cancer per se has been reported in detail within previous publications [46,47]. Here, we discuss the current knowledge about the role of PC-derived exosomes and their cargo in PCRD.

4.1. Exosomal Protein Cargo and PCRD

Studies have shown that certain proteins contained within exosomes derived from pancreatic tumour cells play key roles in the development of PCRD: the most important being adrenomedullin (AM).

Adrenomedullin

This component of PC-derived exosomes is a multifunctional vasoactive peptide that has been implicated in inflammation and sepsis and is overexpressed in PC at both the mRNA and protein levels [48]. High expressions of AM can cause decreased β-cell insulin secretion, as evidenced by studies showing that when β cell lines have been treated with glucose followed by exposure to AM, glucose-stimulated insulin secretion was inhibited [17]. This effect is more pronounced in β-cell lines or islets when isolated from mice but has also been rectified following the small hairpin RNA (shRNA)-mediated knockdown of AM in PC cells. Another study showed that PC-derived exosomes containing AM and carbohydrate antigen 19-9 (CA19-9, also known as sialyl Lewis-a (sLea)—a widely used diagnostic and prognostic marker for PC [49]—readily entered β cells through caveolin-mediated endocytosis or macropinocytosis, and induced cyclic adenosine monophosphate (cAMP) production which is usually associated with increased insulin secretion [42]. However, low insulin secretion was also observed in this study despite the overexpression of cAMP; this points to the induction of endoplasmic reticulum (ER) stress in a process independent of cAMP. Interestingly, the ER stress markers immunoglobulin heavy chain binding protein (BiP) and C/EBP homologous protein (CHOP) were overexpressed, causing an increase in the unfolded protein response (UPR) and overwhelming ER stress in the β cells. This resulted in the upregulation of oxidative stress as indicated by the high expressions of reactive oxygen species (ROS) and reactive nitrogen species (RNS), leading to marked β cell dysfunction. Hence, one of the ways in which PC causes paraneoplastic β cell dysfunction may be through the shedding of AM(+)/CA19-9(+) exosomes into the circulation that inhibit insulin secretion in PCRD; this is likely through the AM-induced failure of UPR and ER stress [42].
Studies in mice have also shown that AM and its receptor components—the calcitonin receptor-like receptor (CRLR) and receptor activity modifying proteins (RAMPs)—are present in pancreatic islets and can be co-localized with insulin [50]. Another AM receptor (ADMR) found on β cells acted as a peptide mediator for AM and was carried within PC-derived exosomes. The interaction of AM with this receptor caused impaired insulin secretion, as seen in PCRD, to initiate the dysfunction of the recipient cells [42]. Interestingly, the ADMR blockade abrogated the inhibitory effect of exosomes during insulin secretion [51] (Figure 3).
Aggarwal et al. demonstrated that mice bearing pancreatic tumours formed from PC cell lines and overexpressing AM were significantly glucose intolerant compared to control mice bearing tumours formed from PC cells transfected with the control vector [17]. In addition to inducing glucose intolerance, PC-derived exosomal AM was found to promote lipolysis in murine and human adipocytes. AM interacted with its receptor on the adipocytes, activated p38, extracellular signal-regulated kinase 1/2 (ERK1/2) and mitogen-activated protein kinases (MAPKs) and promoted lipolysis through the phosphorylation of hormone-sensitive lipase [51]. Hence, AM in pancreatic tumour exosomes may play a role in PCRD not only via its effects on the β cell function but also by inducing lipolysis, which is a major contributor to insulin resistance.
Studies have also identified other proteins which have been speculated to be involved in the pathogenesis of PCRD and which have not yet been identified or characterized as exosomal cargo. These include:
1.
The PC-derived S-100A8 N-terminal peptide: a diabetogenic agent with reduced glucose consumption and lactate production by myoblasts in vitro [52]. This peptide also hindered the growth of myoblasts by inhibiting myotubular differentiation and increasing caspase-3 activation in cultured C2C12 myoblasts. S-100A8 is, therefore, suggested to be a cause of hyperglycemia because it impairs glucose catabolism in myoblasts and is speculated to be a promising biomarker for the diagnosis of PCRD. In addition, S-100A8 N-terminal alters β-cell insulin secretion by inhibiting glucose-stimulated insulin exocytosis (early response), which is characteristic of PCRD [53].
2.
The insulin-like growth factor 1 (IGF-1) and Insulin-like growth factor-binding protein-2 (IGFBP-2) are biomarkers of pancreatic diseases, especially CP and PC. Since the expression of IGF-1 in CP and DM was elevated compared with that in PC and DM, IGF-1 may be an indicator that signals whether pancreatic diabetes is from CP or PC [54].
3.
In the serum taken up to 4 years before a PC diagnosis, circulating Thrombospondin 1 (TSP-1) levels were found to be significantly reduced up to 24 months prior to diagnosis. Low serum TSP-1 levels in PC patients were associated with DM. Interestingly, TSP1 levels in PC patients with DM were lower compared to patients with T2DM alone. TSP-1 was also decreased in PC patients, compared to the healthy controls and patients with benign biliary obstruction at clinical diagnosis. Not only did circulating TSP-1 levels decrease up to 24 months before the diagnosis of PC, but a combination of TSP-1 and CA19-9 also produced a diagnostic capacity that significantly outperformed both markers alone. Decreased TSP-1 levels may be an indication of PCRD, and early PC detection strategies could include exploring the clinical relevance of TSP-1.
4.
The vascular noninflammatory molecule 1 (Vanin 1/VNN1) is a pantetheinase that is anchored to the extracellular membrane of epithelial and myeloid cells. It belongs to an enzymatic pathway and causes oxidative stress, inflammation and cell migration and is suggested to be a biomarker for certain malignancies, including systemic lupus erythematosus and type 1 diabetic nephropathy [55,56]. It was overexpressed in PC and inhibited the growth of insulin-secreting cell lines (β-TC-6 and INS-1) when they were treated with conditioned media derived from PC cell lines. This loss of cell viability was even greater when the cells were exposed to conditioned media from PC cells transfected with a VNN1-overexpressing vector, indicating that the high expression of VNN1 causes injury to paraneoplastic insulin-secreting cells. The high expression of VNN1 also altered the expressions of oxidative stress-related factors, including the downregulation of the anti-oxidative stress/anticancer peroxisome-proliferator activated receptor γ (PPAR-γ), the upregulation of cysteamine which is a product of VNN1, the downregulation of the antioxidant glutathione (GSH) and upregulation of ROS. Hence, the overexpression of VNN1 in PC cells modulated the viability and function of β cells by promoting oxidative stress in the β cells’ microenvironment [57]. Additionally, VNN1, when used in combination with the matrix metalloproteinase 9 (MMP9), could be a more effective blood biomarker panel for the discrimination of PCRD from T2DM [58].

4.2. Exosomal Lipid Cargo and PCRD

There is currently no experimental evidence to show the role of exosomal lipids in PCRD. However, adiponectin, a hormone, which is mainly produced by adipocytes (fat cells), is known to increase insulin sensitivity and regulate peripheral glucose levels and fatty acid metabolism [59]. Additionally, the combination of the adiponectin and interleukin-1 receptor antagonist (IL-1Ra), which was elevated in PC patients with DM compared to those with T2DM and the healthy controls showed strong diagnostic potential for the distinction of PCRD from T2DM [60]. Tumour cells produce a range of interleukins; hence, IL1-R antagonist levels in PC cell-derived exosomes could be worthy of study, particularly in screening for PC in individuals newly diagnosed with T2DM.

4.3. Exosomal RNA Cargo and PCRD

The two types of ribonucleic acid (RNA)—coding and non-coding RNA—are both essential for gene expression. Altered processing or the activity of RNA, whether coding or non-coding, is a hallmark of cancers [61]. However, over the years, there has been accumulated evidence regarding the role of the aberrant expression of non-coding RNAs (ncRNAs) in many cancers [62,63,64,65], including PC and also in other diseases such as DM. These ncRNAs play important roles in epigenetic processes, transcription and post-transcriptional regulation and can, thus, affect the growth, migration and invasion of cells. The role of ncRNAs in cancers (including PC) and other human diseases (including DM) has been reviewed elsewhere [66,67].
Examples of ncRNAs include small RNAs such as microRNAs (miRNAs), small interfering RNAs (siRNAs), Piwi-interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs), small nuclear RNAs (snRNAs), small cajal body-specific RNAs (scaRNAs), as well as extracellular RNAs (exRNAs), long non-coding RNAs (lncRNAs), long intergenic non-coding RNAs (lincRNAs), circular RNAs (cirRNAs), transfer RNAs (tRNAs), messenger RNAs (mRNAs) and ribosomal RNAs (rRNAs). Of these non-coding RNAs, the best studied with regard to exosmal cargo and PCRD are miRNAs, with little information available about other exosomal non-coding RNAs.

Exosomal miRNAs and PCRD

Several studies indicate that microRNA (miRNA) fragments have diagnostic, predictive and therapeutic usefulness in different cancer types, including PC. With regard to PCRD, the effects of exosomes released by PC cells (MIA PaCa-2) on the intestinal secretin tumour cell line (STC-1), which expresses many features of normal enteroendocrine cells, were explored. Normally, enteroendocrine cells are responsible for the production of a glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP-1), both of which function in maintaining glucose homeostasis by enhancing insulin secretion and sensitivity [68]. Zhang et al. found that the treatment of STC-1 cells with exosomes derived from PC cells resulted in the downregulation of GLP-1 and GIP [69]. These authors reported that this effect was mediated by miR-6796-3p, miR-6763-5p, miR-4750-3p and miR-197-3p within the exosomes, as evidenced by the fact that the inhibition of the expression of these miRNAs in the parental tumour cells (MiaPaCa-2) reversed the downregulation of GLP-1 and GIP by downregulating the post-translational proprotein convertase subtilisin/kexin type 1/3 (PCSK1/3) levels in STC-1 cells [69].
In a study by Su et al., exosomal miR-19a from PC cells was found to downregulate insulin secretion in mouse insulinoma 6 cells (MIN6) and primary islets by targeting Neurod1: the validated gene involved in insulin secretion [70]. cAMP, which potentiates insulin secretion, is produced by the cell membrane protein adenylyl cyclase (ADCY) and ATP, and Adcy1 is a member of the ADCY family, which is activated by extracellular Ca2+ in MIN6 cells [71]. The binding of miR-19a to Neurod1 caused the downregulation of cAMP and Ca2+ expression in β cells, leading to decreased insulin secretion [70]. Another independent report demonstrated that PC-derived exosomal miR-19a targets Adcy1 and Epac2 were an exchange protein directly activated by cAMP 2. Both these factors are involved in insulin secretion, thereby inducing β cell dysfunction in PCRD [43]. This indicates that signal changes between PC cells and β cells via exosomes might be important in the pathogenesis of PCRD and provide a possible basis for the application of exosomal miR-19a in a PC screening strategy.
Regarding insulin resistance, PC-derived exosomes were found to readily enter C2C12 myotubes, causing lipidosis and the inhibition of glucose uptake by impairing glucose transporter type 4 (GLUT4) trafficking and preserving forkhead box protein O1 (FOXO1) nuclear exclusion [72]. FOXO1 is a nuclear transcription factor whose expression can be induced by the presence of insulin; therefore, it can be overexpressed in skeletal muscles in energy-deprived states such as diabetes and is a key target of the PI3K/Akt signalling pathway [73]. Once activated/phosphorylated by activated Akt, FOXO1 could be excluded from the nucleus, resulting in the repression of transcription and muscle insulin resistance. FOXO1 in PC-derived exosomes inhibited GLUT4 expression as well as insulin and PI3K/Akt signalling [72]. Interestingly, miRNAs were found to be upregulated in PC-derived exosomes including miR-666-3p, miR-540-3p, miR-125b-5p and miR-450b-3p promoted FOXO1 expression, while miR-883b-5p, 666-3p, miR-450b-3p and miR-151-3p downregulated GLUT4 expression. Therefore, PC-derived exosomal miRNAs were found to be capable of inducing the insulin resistance of skeletal muscle cells through insulin and PI3K/Akt/FoxO1 signalling pathways [72].
Another miRNA that could play a role in insulin resistance is miRNA-let-7b-5p, which has been shown to interact directly with SLC6A15: a gene associated with the insulin resistance of metabolic disorders. miRNA-let-7b-5p inhibits SLC6A15 expression via its effects on the SLC6A15 3′-untranslated region (3′-UTR) and is thought to promote insulin resistance in C2C12 myotube cells [74].
Studies have also shown a few other non-coding RNAs, including miRNAs and long non-coding RNAs (lncRNAs), which may be involved in the pathogenesis of PCRD but are yet to be investigated in exosomes. For instance, a panel of six serum miRNAs (miR-483-5p, miR-19a, miR-29a, miR-20a, miR-24, miR-25) upregulated in PCRD was developed as a biomarker for PCRD, with a significant diagnostic efficacy for the discrimination of PCRD from non-cancer new-onset T2DM, signifying that they may be involved in PCRD development [75].
Another study reported the association of seven lncRNAs and two miRNAs with prognosis in patients with PCRD. The lncRNAs included (i) the DiGeorge syndrome critical region gene 9 (DGCR9), (ii) FLJ33360, (iii) H19, (iv) HOX Transcript Antisense RNA (HOTAIR), (v) KIAA0125, (vi) Small Cajal Body-Specific RNA 2 (SCARNA2) and (vii) Urothelial Cancer Associated 1 (UCA1). The two miRNAs were (i) hsa-miR-214 and (ii) hsa-miR-429. Notably, hsa-miR-214 was predicted to target the lncRNA HOTAIR and its downstream mRNA target Coiled-Coil Domain Containing 33 (CCDC33). Similarly, hsa-miR-429 could target Cat Eye Syndrome Chromosome Region, Candidate 7 (CECR7) and the Cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4) downstream, suggesting that these RNAs regulate each other post-transcriptionally as they compete when binding to the miRNAs (a phenomenon known as the competing endogenous RNA network). ‘HOTAIR-hsa-miR-214-CCDC33’ and ‘CECR7-hsa-miR-429-CTLA4’ regulations might be two important mechanisms for PC progression, requiring further investigation. HOTAIR, CECR7, UCA1, hsa-miR-214, hsa-miR-429, CCDC33 and CTLA4 were also highlighted as potential diagnostic biomarkers [76], though these were not characterized in exosomes.

5. Tumour-Derived Exosomal Cargo and Pancreatic Cancer

Additionally, worthy of note is the fact that exosomes derived from pancreatic tumours can exacerbate the aggressiveness of PC mainly via their protein and RNA cargo components [77]. By taking up exosomes/exosomal cargo released by highly malignant PC cell lines, moderately malignant PC cell lines acquire increased proliferative, migratory, and invasive abilities. For example, the zinc/iron-regulated transporter-like protein 4 (ZIP4) is a protein upregulated in the highly malignant PC cell line—PC-1.0 that promotes tumour growth both in vitro and in vivo [78]. When less malignant PC cell lines such as PC-1 take up ZIP4-containing exosomes derived from PC-1.0 cells, the recipient cells become more aggressive, suggesting the role of exosomal ZIP4 in PC progression. Exosomal proteins have also been reported to be associated with the resistance of PC cells to gemcitabine treatment. Exosomes derived from the PANC-1 cell line (which is more prone to gemcitabine resistance) were shown to overexpress the Ephrin type-A receptor 2 (EphA2) compared with other less chemoresistant cell lines (MIA PaCa-2 and BxPC-3) [79]. When EphA2 expression was suppressed in PANC-1 cells, their ability to transmit exosome-mediated chemoresistance to MIA PaCa-2 and BxPC-3 cells was inhibited, suggesting the possible role of EphA2 in promoting the resistance of PANC-1 cells to gemcitabine treatment and the transmission of the same to other cells.
Regarding RNAs, many studies have shown how RNAs, especially non-coding RNAs contribute to PC development and progression. For instance, miR-301a-3p derived from hypoxic PC cells induced the differentiation of macrophages into the M2 phenotype and enhanced the metastatic competence of PC cells by activating the PTEN/PI3Kγ signalling pathway [80]. Similarly, exosomal LINC01133 was found to be overexpressed in PC and associated with disease progression and metastasis [81]. LINC01133 interacted with the enhancer of zeste homolog 2 (EZH2) to promote the trimethylation of histone H3 on lysine 27 (H3K27). This was achieved via the suppression of the axis inhibition protein 2 (AXIN2) and glycogen synthase kinase 3 (GSK-3) activities, resulting in the activation of β-catenin. This promoted an epithelial-mesenchymal transition (EMT). These and many other studies [82,83,84,85] have proven that exosomal RNAs can be associated with pancreatic malignancy.

6. Pancreatic Stellate Cell-Derived Exosomes and PCRD

Pancreatic stellate cells (PSCs) are stromal cells that are present around the earliest neoplastic lesions and around islets. PSCs are responsible for producing the collagenous stroma of PC and interact closely with cancer cells to drive cancer progression. Recent studies suggest that exosomes secreted by PSCs may play a major role in stroma-tumour cross-talk [86].
In this regard, exosomal miR-5703 was found to target CKLF Like MARVEL Transmembrane Domain Containing 4 (CMTM4) which is known to regulate the expression of the inhibitory programmed death-ligand 1 (PD-L1) and to stabilize the PD-L1 receptor in cancers. By binding to CMTM4, the PC-derived exosomal miR-5703 promoted the proliferation of PC cells via the PAK4-activated PI3K/Akt pathway [87]. Additionally, when PC cells internalized PSC-derived exosomes, this resulted in high miR-21 levels, which promoted cell migration, EMT, increased matrix metalloproteinase-2/9 activity and enhanced Ras/ERK signalling activity by increasing ERK1/2 and Akt phosphorylation in the PC cells [88]. Additionally, hypoxia-induced PSC-derived exosomal miRNAs—miR-4465 and miR-616-3p—targeted the tumour-suppressing phosphatase and tensin homolog (PTEN) and activated AKT signalling in PC cells, promoting proliferation, migration and invasion [89].
Of particular relevance to this review is a recent study that demonstrated how exosomes derived from co-cultures of mouse PSCs and PC cells caused mouse β cell dysfunction by decreasing the β cell proliferation and inhibiting their insulin secretion capacity in response to glucose stimulation, showing that exosomes secreted by PSCs and PC cells carry factors that negatively regulate the functions of islet cells [90] (Figure 4). Whether PSC-derived exosomes play a role in peripheral insulin resistance is yet to be determined. Nevertheless, it is possible that the exosomal cargo from both PC cells and stromal cells (PSCs) play a role in the pathogenesis of PCRD.

7. Other Biomarkers for PCRD

Studies have also identified some mechanisms and genomic markers for PCRD, although the localization of these biomarkers in exosomes has not yet been established. For example:
1.
Nitric oxide (NO), which is overexpressed in PCRD, has diabetogenic effects, and the absence of specific fragments of NO correlates with the development of DM [91]. Hence, PCRD might be a result of the diabetogenic effects of tumour products, possibly acting via NO.
2.
In a study aimed at identifying the mediators of PCRD by comparing the gene expression between PCRD patients and PC patients without DM, low levels of Kinesin Family Member 22 (KIF22) and glycogen Phosphorylase L (PYGL) were found to be associated with good survival outcomes for PC patients with DM and may be prognostic biomarkers for PCRD. Additionally, bioinformatic predictions revealed that KIF22, PYGL, Ribosomal Protein S27a (RPS27A), and ubiquitin A-52 residue ribosomal protein fusion product 1 (UBA52) could be involved in the pathogenesis of PCRD [92].
Further studies are required to determine whether the role of these markers in PCRD is exosome-based. Considering the stability, relevance, and reliability of exosome-based biomarkers, we recommend that in future studies, the functions of these biomarkers are examined to determine whether they are enhanced by exosomes.

8. Summary

Pancreatic cancer-related diabetes (PCRD) is a condition that identifies a specific subset of patients who may feasibly benefit from the screening strategies for pancreatic cancer. Understanding the pathogenesis of PCRD, particularly the role of pancreatic cancer-derived exosomal cargo in disrupting glucose homeostasis is an important first step when identifying the factors that could be used as biomarkers for early diagnosis, the prediction of response to therapy as well as prognosis. The role of PC-derived exosomal cargo components, particularly proteins and microRNAs, in PCRD, is becoming increasingly elucidated. Additionally, the exosomal factors involved in PCRD but not longstanding Type 2 diabetes have also been identified. Thus, there are some promising advances in this field that bode well for the development of clinical applications in the near future and could enable timely diagnosis and improved outcomes for patients with pancreatic cancer.

Author Contributions

H.B.B. searched the literature, wrote the manuscript, and prepared the table and figures. C.J.P. and M.V.A. conceived, revised, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This was supported by the Ideas Grant (RG191448) received from the National Health and Medical Research Council, Australia, and the Pankind Early Detection Innovation Grant (RG213683) received from the Pankind, The Australian Pancreatic Cancer Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Olusanya Olasehinde for assisting with the design of the figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sarantis, P.; Koustas, E.; Papadimitropoulou, A.; Papavassiliou, A.G.; Karamouzis, M.V. Pancreatic ductal adenocarcinoma: Treatment hurdles, tumor microenvironment and immunotherapy. World J. Gastrointest. Oncol. 2020, 12, 173–181. [Google Scholar] [CrossRef] [PubMed]
  2. Hackeng, W.M.; Hruban, R.H.; Offerhaus, G.J.A.; Brosens, L.A.A. Surgical and molecular pathology of pancreatic neoplasms. Diagn. Pathol. 2016, 11, 47. [Google Scholar] [CrossRef] [Green Version]
  3. Wang, H.; Liu, J.; Xia, G.; Lei, S.; Huang, X.; Huang, X. Survival of pancreatic cancer patients is negatively correlated with age at diagnosis: A population-based retrospective study. Sci. Rep. 2020, 10, 7048. [Google Scholar] [CrossRef]
  4. Australian Institute of Health and Welfare. Cancer Data in Australia [Internet]. Canberra: Australian Institute of Health and Welfare. 2022. Available online: https://www.aihw.gov.au/reports/cancer/cancer-data-in-australia (accessed on 27 April 2023).
  5. Khadka, R.; Tian, W.J.; Hao, X.; Koirala, R. Risk factor, early diagnosis and overall survival on outcome of association between pancreatic cancer and diabetes mellitus: Changes and advances, a review. Int. J. Surg. 2018, 52, 342–346. [Google Scholar] [CrossRef] [PubMed]
  6. Capasso, M.; Franceschi, M.; Rodriguez-Castro, K.I.; Crafa, P.; Cambiè, G.; Miraglia, C.; Barchi, A.; Nouvenne, A.; Leandro, G.; Meschi, T.; et al. Epidemiology and risk factors of pancreatic cancer. Acta Bio Med. Atenei Parm. 2018, 89, 141–146. [Google Scholar] [CrossRef]
  7. Lam, B.Q.; Shrivastava, S.K.; Shrivastava, A.; Shankar, S.; Srivastava, R.K. The Impact of obesity and diabetes mellitus on pancreatic cancer: Molecular mechanisms and clinical perspectives. J. Cell. Mol. Med. 2020, 24, 7706–7716. [Google Scholar] [CrossRef]
  8. Malka, D.; Hammel, P.; Maire, F.; Rufat, P.; Madeira, I.; Pessione, F.; Lévy, P.; Ruszniewski, P. Risk of pancreatic adenocarcinoma in chronic pancreatitis. Gut 2002, 51, 849–852. [Google Scholar] [CrossRef] [PubMed]
  9. Pothuraju, R.; Rachagani, S.; Junker, W.M.; Chaudhary, S.; Saraswathi, V.; Kaur, S.; Batra, S.K. Pancreatic cancer associated with obesity and diabetes: An alternative approach for its targeting. J. Exp. Clin. Cancer Res. 2018, 37, 319. [Google Scholar] [CrossRef] [Green Version]
  10. Ewald, N. Diagnosis and treatment of diabetes mellitus in chronic pancreatitis. World J. Gastroenterol. 2013, 19, 7276. [Google Scholar] [CrossRef]
  11. Illés, D.; Ivány, E.; Holzinger, G.; Kosár, K.; Adam, M.G.; Kamlage, B.; Zsóri, G.; Tajti, M.; Svébis, M.M.; Horváth, V.; et al. New Onset of DiabetEs in aSsociation with pancreatic ductal adenocarcinoma (NODES Trial): Protocol of a prospective, multicentre observational trial. BMJ Open 2020, 10, e037267. [Google Scholar] [CrossRef]
  12. Khan, M.A.B.; Hashim, M.J.; King, J.K.; Govender, R.D.; Mustafa, H.; Al Kaabi, J. Epidemiology of Type 2 Diabetes–Global Burden of Disease and Forecasted Trends. J. Epidemiol. Glob. Health 2020, 10, 107–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Patel, R.; Ede, J.; Collins, J.; Willens, D. Pancreatic Cancer Presenting as New-Onset Diabetes. Case Rep. Oncol. 2014, 7, 171–174. [Google Scholar] [CrossRef] [PubMed]
  14. Mellenthin, C.; Balaban, V.D.; Dugic, A.; Cullati, S. Risk Factors for Pancreatic Cancer in Patients with New-Onset Diabetes: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 4684. [Google Scholar] [CrossRef] [PubMed]
  15. Parajuli, P.; Nguyen, T.L.; Prunier, C.; Razzaque, M.S.; Xu, K.; Atfi, A. Pancreatic cancer triggers diabetes through TGF-β–mediated selective depletion of islet β-cells. Life Sci. Alliance 2020, 3, e201900573. [Google Scholar] [CrossRef]
  16. Andersen, D.K.; Andren-Sandberg, Å.; Duell, E.J.; Goggins, M.; Korc, M.; Petersen, G.M.; Smith, J.P.; Whitcomb, D.C. Pancreatitis-Diabetes-Pancreatic Cancer: Summary of an NIDDK-NCI Workshop. Pancreas 2013, 42, 1227–1237. [Google Scholar] [CrossRef] [Green Version]
  17. Aggarwal, G.; Ramachandran, V.; Javeed, N.; Arumugam, T.; Dutta, S.; Klee, G.G.; Klee, E.W.; Smyrk, T.C.; Bamlet, W.; Han, J.J.; et al. Adrenomedullin is Up-regulated in Patients with Pancreatic Cancer and Causes Insulin Resistance in β Cells and Mice. Gastroenterology 2012, 143, 1510.e1–1517.e1. [Google Scholar] [CrossRef] [Green Version]
  18. Vázquez, R.; Riveiro, M.E.; Berenguer-Daizé, C.; O’kane, A.; Gormley, J.; Touzelet, O.; Rezai, K.; Bekradda, M.; Ouafik, L. Targeting Adrenomedullin in Oncology: A Feasible Strategy with Potential as Much More Than an Alternative Anti-Angiogenic Therapy. Front. Oncol. 2021, 10, 589218. [Google Scholar] [CrossRef]
  19. Keleg, S.; Kayed, H.; Jiang, X.; Penzel, R.; Giese, T.; Büchler, M.W.; Friess, H.; Kleeff, J. Adrenomedullin is induced by hypoxia and enhances pancreatic cancer cell invasion. Int. J. Cancer 2007, 121, 21–32. [Google Scholar] [CrossRef]
  20. Permert, J.; Larsson, J.; Westermark, G.T.; Herrington, M.K.; Christmanson, L.; Pour, P.M.; Westermark, P.; Adrian, T.E. Islet Amyloid Polypeptide in Patients with Pancreatic Cancer and Diabetes. N. Engl. J. Med. 1994, 330, 313–318. [Google Scholar] [CrossRef]
  21. Hart, P.A.; Baichoo, E.; Bi, Y.; Hinton, A.; Kudva, Y.C.; Chari, S.T. Pancreatic polypeptide response to a mixed meal is blunted in pancreatic head cancer associated with diabetes mellitus. Pancreatology 2015, 15, 162–166. [Google Scholar] [CrossRef]
  22. Narasimhan, A.; Zhong, X.; Au, E.P.; Ceppa, E.P.; Nakeeb, A.; House, M.G.; Zyromski, N.J.; Schmidt, C.M.; Schloss, K.N.H.; Schloss, D.E.I.; et al. Profiling of Adipose and Skeletal Muscle in Human Pancreatic Cancer Cachexia Reveals Distinct Gene Profiles with Convergent Pathways. Cancers 2021, 13, 1975. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, P.C.; You, M.; Yu, S.-Y.; Luan, Y.; Eldani, M.; Caffrey, T.C.; Grandgenett, P.M.; O’connell, K.A.; Shukla, S.K.; Kattamuri, C.; et al. Visceral adipose tissue remodeling in pancreatic ductal adenocarcinoma cachexia: The role of activin A signaling. Sci. Rep. 2022, 12, 1659. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, Y.; Kim, O.-K. Potential Roles of Adipocyte Extracellular Vesicle–Derived miRNAs in Obesity-Mediated Insulin Resistance. Adv. Nutr. Int. Rev. J. 2021, 12, 566–574. [Google Scholar] [CrossRef]
  25. Danai, L.V.; Babic, A.; Rosenthal, M.H.; Dennstedt, E.A.; Muir, A.; Lien, E.C.; Mayers, J.R.; Tai, K.; Lau, A.N.; Jones-Sali, P.; et al. Altered exocrine function can drive adipose wasting in early pancreatic cancer. Nature 2018, 558, 600–604. [Google Scholar] [CrossRef] [PubMed]
  26. Aly, H.F. How pancreatic adenocarcinoma might cause diabetes? The role of TGF-β. Int. J. Clin. Gastroenterol. Hepatol. 2021, 3, 5–10. [Google Scholar]
  27. Motohashi, N.; Alexander, M.S.; Shimizu-Motohashi, Y.; Myers, J.A.; Kawahara, G.; Kunkel, L.M. Regulation of IRS1/Akt insulin signaling by microRNA-128a during myogenesis. J. Cell Sci. 2013, 126, 2678–2691. [Google Scholar] [CrossRef] [Green Version]
  28. He, J.; Li, Y.; Yang, X.; He, X.; Zhang, H.; Zhang, L.; He, J. The Feedback Regulation of PI3K-miR-19a, and MAPK-miR-23b/27b in Endothelial Cells under Shear Stress. Molecules 2012, 18, 1–13. [Google Scholar] [CrossRef] [Green Version]
  29. Roy, A.; Sahoo, J.; Kamalanathan, S.; Naik, D.; Mohan, P.; Kalayarasan, R. Diabetes and pancreatic cancer: Exploring the two-way traffic. World J. Gastroenterol. 2021, 27, 4939–4962. [Google Scholar] [CrossRef]
  30. De Souza, A.; Irfan, K.; Masud, F.; Saif, M.W. Diabetes Type 2 and Pancreatic Cancer: A History Unfolding. JOP J. Pancreas 2016, 17, 144. [Google Scholar]
  31. Albury-Warren, T. Diabetes Phenotypes in Transgenic Pancreatic Cancer Mouse Models. Ph.D. Thesis, University of Central Florida, Orlando, FL, USA, 2015. [Google Scholar]
  32. Gupta, S.; Vittinghoff, E.; Bertenthal, D.; Corley, D.; Shen, H.; Walter, L.C.; McQuaid, K. New-Onset Diabetes and Pancreatic Cancer. Clin. Gastroenterol. Hepatol. 2006, 4, 1366–1372. [Google Scholar] [CrossRef]
  33. Gallo, M.; Adinolfi, V.; Morviducci, L.; Acquati, S.; Tuveri, E.; Ferrari, P.; Zatelli, M.; Faggiano, A.; Argentiero, A.; Natalicchio, A.; et al. Early prediction of pancreatic cancer from new-onset diabetes: An Associazione Italiana Oncologia Medica (AIOM)/Associazione Medici Diabetologi (AMD)/Società Italiana Endocrinologia (SIE)/Società Italiana Farmacologia (SIF) multidisciplinary consensus position paper. ESMO Open 2021, 6, 100155. [Google Scholar] [CrossRef]
  34. Lazo, S.; Hooten, N.N.; Green, J.; Eitan, E.; Mode, N.A.; Liu, Q.; Zonderman, A.B.; Ezike, N.; Mattson, M.P.; Ghosh, P.; et al. Mitochondrial DNA in extracellular vesicles declines with age. Aging Cell 2020, 20, e13283. [Google Scholar] [CrossRef]
  35. Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  36. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef] [PubMed]
  38. Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [Green Version]
  39. Wu, S.F.; Noren Hooten, N.; Freeman, D.W.; Mode, N.A.; Zonderman, A.B.; Evans, M.K. Extracellular vesicles in diabetes mellitus induce alterations in endothelial cell morphology and migration. J. Transl. Med. 2020, 18, 230. [Google Scholar] [CrossRef] [PubMed]
  40. Xiao, Y.; Zheng, L.; Zou, X.; Wang, J.; Zhong, J.; Zhong, T. Extracellular vesicles in type 2 diabetes mellitus: Key roles in pathogenesis, complications, and therapy. J. Extracell. Vesicles 2019, 8, 1625677. [Google Scholar] [CrossRef] [Green Version]
  41. Moeng, S.; Son, S.W.; Lee, J.S.; Lee, H.Y.; Kim, T.H.; Choi, S.Y.; Kuh, H.J.; Park, J.K. Extracellular Vesicles (EVs) and Pancreatic Cancer: From the Role of EVs to the Interference with EV-Mediated Reciprocal Communication. Biomedicines 2020, 8, 267. [Google Scholar] [CrossRef]
  42. Javeed, N.; Sagar, G.; Dutta, S.K.; Smyrk, T.C.; Lau, J.S.; Bhattacharya, S.; Truty, M.; Petersen, G.M.; Kaufman, R.J.; Chari, S.T.; et al. Pancreatic Cancer–Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin. Cancer Res. 2015, 21, 1722–1733. [Google Scholar] [CrossRef] [Green Version]
  43. Pang, W.; Yao, W.; Dai, X.; Zhang, A.; Hou, L.; Wang, L.; Wang, Y.; Huang, X.; Meng, X.; Li, L. Pancreatic cancer-derived exosomal microRNA-19a induces β-cell dysfunction by targeting ADCY1 and EPAC2. Int. J. Biol. Sci. 2021, 17, 3622–3633. [Google Scholar] [CrossRef] [PubMed]
  44. Halvaei, S.; Daryani, S.; Eslami-S, Z.; Samadi, T.; Jafarbeik-Iravani, N.; Bakhshayesh, T.O.; Majidzadeh-A, K.; Esmaeili, R. Exosomes in Cancer Liquid Biopsy: A Focus on Breast Cancer. Mol. Ther. Nucleic Acids 2017, 10, 131–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Verel-Yilmaz, Y.; Fernández, J.P.; Schäfer, A.; Nevermann, S.; Cook, L.; Gercke, N.; Helmprobst, F.; Jaworek, C.; von Strandmann, E.P.; Pagenstecher, A.; et al. Extracellular Vesicle-Based Detection of Pancreatic Cancer. Front. Cell Dev. Biol. 2021, 9, 697939. [Google Scholar] [CrossRef] [PubMed]
  46. Chang, C.-H.; Pauklin, S. Extracellular vesicles in pancreatic cancer progression and therapies. Cell Death Dis. 2021, 12, 973. [Google Scholar] [CrossRef]
  47. Channon, L.M.; Tyma, V.M.; Xu, Z.; Greening, D.W.; Wilson, J.S.; Perera, C.J.; Apte, M.V. Small extracellular vesicles (exosomes) and their cargo in pancreatic cancer: Key roles in the hallmarks of cancer. Biochim. Biophys. Acta (BBA) Rev. Cancer 2022, 1877, 188728. [Google Scholar] [CrossRef] [PubMed]
  48. Andersen, D.K.; Korc, M.; Petersen, G.M.; Eibl, G.; Li, D.; Rickels, M.R.; Chari, S.T.; Abbruzzese, J.L. Diabetes, Pancreatogenic Diabetes, and Pancreatic Cancer. Diabetes 2017, 66, 1103–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ballehaninna, U.K.; Chamberlain, R.S. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. J. Gastrointest. Oncol. 2012, 3, 105–119. [Google Scholar] [CrossRef]
  50. Cueille, C.; Birot, O.; Bigard, X.; Hagner, S.; Garel, J.-M. Post-transcriptional regulation of CRLR expression during hypoxia. Biochem. Biophys. Res. Commun. 2004, 326, 23–29. [Google Scholar] [CrossRef]
  51. Sagar, G.; Sah, R.P.; Javeed, N.; Dutta, S.K.; Smyrk, T.C.; Lau, J.S.; Giorgadze, N.; Tchkonia, T.; Kirkland, J.L.; Chari, S.T.; et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut 2015, 65, 1165–1174. [Google Scholar] [CrossRef] [Green Version]
  52. Basso, D.; Greco, E.; Fogar, P.; Pucci, P.; Flagiello, A.; Baldo, G.; Giunco, S.; Valerio, A.; Navaglia, F.; Zambon, C.-F.; et al. Pancreatic cancer-derived S-100A8 N-terminal peptide: A diabetes cause? Clin. Chim. Acta 2006, 372, 120–128. [Google Scholar] [CrossRef]
  53. Padoan, A.; Greco, E.; Basso, D.; Fogar, P.; Valerio, A.N.N.A.; Fadi, E.; Schiavon, S.; Fasolo, M.; Vigolo, S.; DE CARLO, E.; et al. The pancreatic cancer derived N-Terminal peptide of S100A8 inhibits insulin exocytosis and stimulates cancer cell growth. Biochim. Clin. 2007, 31, 432. [Google Scholar]
  54. Włodarczyk, B.; Borkowska, A.; Włodarczyk, P.; Małecka-Panas, E.; Gąsiorowska, A. Insulin-like growth factor 1 and insulin-like growth factor binding protein 2 serum levels as potential biomarkers in differential diagnosis between chronic pancreatitis and pancreatic adenocarcinoma in reference to pancreatic diabetes. Gastroenterol. Rev. 2021, 16, 36–42. [Google Scholar] [CrossRef]
  55. Sánchez-Muñoz, F.; Amezcua-Guerra, L.; Macías-Palacios, M.; Márquez-Velasco, R.; Bojalil, R. Vanin-1 as a potential novel biomarker for active nephritis in systemic lupus erythematosus. Lupus 2013, 22, 333–335. [Google Scholar] [CrossRef]
  56. Fugmann, T.; Borgia, B.; Révész, C.; Godó, M.; Forsblom, C.; Hamar, P.; Holthöfer, H.; Neri, D.; Roesli, C. Proteomic identification of vanin-1 as a marker of kidney damage in a rat model of type 1 diabetic nephropathy. Kidney Int. 2011, 80, 272–281. [Google Scholar] [CrossRef] [Green Version]
  57. Kang, M.; Qin, W.; Buya, M.; Dong, X.; Zheng, W.; Lu, W.; Chen, J.; Guo, Q.; Wu, Y. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett. 2016, 373, 241–250. [Google Scholar] [CrossRef]
  58. Huang, H.; Dong, X.; Kang, M.X.; Xu, B.; Chen, Y.; Zhang, B.; Chen, J.; Xie, Q.P.; Wu, Y.L. Novel Blood Biomarkers of Pancreatic Cancer–Associated Diabetes Mellitus Identified by Peripheral Blood–Based Gene Expression Profiles. Am. J. Gastroenterol. 2010, 105, 1661–1669. [Google Scholar] [CrossRef]
  59. Fisman, E.Z.; Tenenbaum, A. Adiponectin: A manifold therapeutic target for metabolic syndrome, diabetes, and coronary disease? Cardiovasc. Diabetol. 2014, 13, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Oldfield, L.; Evans, A.; Rao, R.G.; Jenkinson, C.; Purewal, T.; Psarelli, E.E.; Menon, U.; Timms, J.F.; Pereira, S.P.; Ghaneh, P.; et al. Blood levels of adiponectin and IL-1Ra distinguish type 3c from type 2 diabetes: Implications for earlier pancreatic cancer detection in new-onset diabetes. Ebiomedicine 2022, 75, 103802. [Google Scholar] [CrossRef] [PubMed]
  61. Goodall, G.J.; Wickramasinghe, V.O. RNA in cancer. Nat. Rev. Cancer 2021, 21, 22–36. [Google Scholar] [CrossRef] [PubMed]
  62. Binang, H.B.; Wang, Y.; Tewara, M.A.; Du, L.; Shi, S.; Li, N.; Nsenga, A.G.A.; Wang, C. Expression levels and associations of five long non-coding RNAs in gastric cancer and their clinical significance. Oncol. Lett. 2020, 19, 2431–2445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  64. Le, P.; Romano, G.; Nana-Sinkam, P.; Acunzo, M. Non-Coding RNAs in Cancer Diagnosis and Therapy: Focus on Lung Cancer. Cancers 2021, 13, 1372. [Google Scholar] [CrossRef] [PubMed]
  65. Qian, Y.; Shi, L.; Luo, Z. Long Non-coding RNAs in Cancer: Implications for Diagnosis, Prognosis, and Therapy. Front. Med. 2020, 7, 902. [Google Scholar] [CrossRef]
  66. Chen, F.; Wang, N.; Tan, H.-Y.; Guo, W.; Zhang, C.; Feng, Y. The functional roles of exosomes-derived long non-coding RNA in human cancer. Cancer Biol. Ther. 2019, 20, 583–592. [Google Scholar] [CrossRef]
  67. Li, C.; Ni, Y.-Q.; Xu, H.; Xiang, Q.-Y.; Zhao, Y.; Zhan, J.-K.; He, J.-Y.; Li, S.; Liu, Y.-S. Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct. Target. Ther. 2021, 6, 383. [Google Scholar] [CrossRef] [PubMed]
  68. Verhoeckx, K.; Cotter, P.; López-Expósito, I.; Kleiveland, C.; Lea, T.; Mackie, A.; Requena, T.; Swiatecka, D.; Wichers, H. (Eds.) The Impact of Food Bioactives on Health; Springer International Publishing: Cham, Switzerland, 2015; Available online: http://link.springer.com/10.1007/978-3-319-16104-4 (accessed on 24 April 2023).
  69. Zhang, Y.; Huang, S.; Li, P.; Chen, Q.; Li, Y.; Zhou, Y.; Wang, L.; Kang, M.; Zhang, B.; Yang, B.; et al. Pancreatic cancer-derived exosomes suppress the production of GIP and GLP-1 from STC-1 cells in vitro by down-regulating the PCSK1/3. Cancer Lett. 2018, 431, 190–200. [Google Scholar] [CrossRef] [PubMed]
  70. Su, J.; Pang, W.; Zhang, A.; Li, L.; Yao, W.; Dai, X. Exosomal miR-19a decreases insulin production by targeting Neurod1 in pancreatic cancer associated diabetes. Mol. Biol. Rep. 2021, 49, 1711–1720. [Google Scholar] [CrossRef] [PubMed]
  71. Kitaguchi, T.; Oya, M.; Wada, Y.; Tsuboi, T.; Miyawaki, A. Extracellular calcium influx activates adenylate cyclase 1 and potentiates insulin secretion in MIN6 cells. Biochem. J. 2013, 450, 365–373. [Google Scholar] [CrossRef] [Green Version]
  72. Wang, L.; Zhang, B.; Zheng, W.; Kang, M.; Chen, Q.; Qin, W.; Li, C.; Zhang, Y.; Shao, Y.; Wu, Y. Exosomes derived from pancreatic cancer cells induce insulin resistance in C2C12 myotube cells through the PI3K/Akt/FoxO1 pathway. Sci. Rep. 2017, 7, 538. [Google Scholar] [CrossRef] [Green Version]
  73. Kamei, Y.; Miura, S.; Suzuki, M.; Kai, Y.; Mizukami, J.; Taniguchi, T.; Mochida, K.; Hata, T.; Matsuda, J.; Aburatani, H.; et al. Skeletal Muscle FOXO1 (FKHR) Transgenic Mice Have Less Skeletal Muscle Mass, Down-regulated Type I (Slow Twitch/Red Muscle) Fiber Genes, and Impaired Glycemic Control. J. Biol. Chem. 2004, 279, 41114–41123. [Google Scholar] [CrossRef] [Green Version]
  74. Wang, L. Exosomal microRNA-let-7b-5p Derived from Pancreatic Cancer Cells Possibly Promotes Insulin Resistance in C2C12 Myotube Cells by Targeting SLC6A15. In Review. 2021. Available online: https://www.researchsquare.com/article/rs-689703/v1 (accessed on 5 August 2022).
  75. Dai, X.; Pang, W.; Zhou, Y.; Yao, W.; Xia, L.; Wang, C.; Chen, X.; Zen, K.; Zhang, C.-Y.; Yuan, Y. Altered profile of serum microRNAs in pancreatic cancer-associated new-onset diabetes mellitus. J. Diabetes 2016, 8, 422–433. [Google Scholar] [CrossRef]
  76. Yao, K.; Wang, Q.; Jia, J.; Zhao, H. A competing endogenous RNA network identifies novel mRNA, miRNA and lncRNA markers for the prognosis of diabetic pancreatic cancer. Tumor Biol. 2017, 39, 1010428317707882. [Google Scholar] [CrossRef] [Green Version]
  77. Lan, B.; Zeng, S.; Grützmann, R.; Pilarsky, C. The Role of Exosomes in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 4332. [Google Scholar] [CrossRef] [Green Version]
  78. Jin, H.; Liu, P.; Wu, Y.; Meng, X.; Wu, M.; Han, J.; Tan, X. Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 2018, 109, 2946–2956. [Google Scholar] [CrossRef] [Green Version]
  79. Fan, J.; Wei, Q.; Koay, E.J.; Liu, Y.; Ning, B.; Bernard, P.W.; Zhang, N.; Han, H.; Katz, M.H.; Zhao, Z.; et al. Chemoresistance Transmission via Exosome-Mediated EphA2 Transfer in Pancreatic Cancer. Theranostics 2018, 8, 5986–5994. [Google Scholar] [CrossRef]
  80. Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kγ to Promote Pancreatic Cancer Metastasis. Cancer Res. 2018, 78, 4586–4598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Liu, Y.; Tang, T.; Yang, X.; Qin, P.; Wang, P.; Zhang, H.; Bai, M.; Wu, R.; Li, F. Tumor-derived exosomal long noncoding RNA LINC01133, regulated by Periostin, contributes to pancreatic ductal adenocarcinoma epithelial-mesenchymal transition through the Wntβ-catenin pathway by silencing AXIN2. Oncogene 2021, 40, 3164–3179. [Google Scholar] [CrossRef] [PubMed]
  82. Robless, E.E.; Howard, J.A.; Casari, I.; Falasca, M. Exosomal long non-coding RNAs in the diagnosis and oncogenesis of pancreatic cancer. Cancer Lett. 2021, 501, 55–65. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, Z.; Zhao, G.; Yang, S.; Zhu, S.; Zhang, S.; Li, P. The significance of exosomal RNAs in the development, diagnosis, and treatment of pancreatic cancer. Cancer Cell Int. 2021, 21, 364. [Google Scholar] [CrossRef] [PubMed]
  84. Reese, M.; Dhayat, S.A. Small extracellular vesicle non-coding RNAs in pancreatic cancer: Molecular mechanisms and clinical implications. J. Hematol. Oncol. 2021, 14, 141. [Google Scholar] [CrossRef] [PubMed]
  85. Sun, C.; Wang, P.; Dong, W.; Liu, H.; Sun, J.; Zhao, L. LncRNA PVT1 promotes exosome secretion through YKT6, RAB7, and VAMP3 in pancreatic cancer. Aging 2020, 12, 10427–10440. [Google Scholar] [CrossRef] [PubMed]
  86. Perera, C.J.; Falasca, M.; Chari, S.T.; Greenfield, J.R.; Xu, Z.; Pirola, R.C.; Wilson, J.S.; Apte, M.V. Role of Pancreatic Stellate Cell-Derived Exosomes in Pancreatic Cancer-Related Diabetes: A Novel Hypothesis. Cancers 2021, 13, 5224. [Google Scholar] [CrossRef]
  87. Li, M.; Guo, H.; Wang, Q.; Chen, K.; Marko, K.; Tian, X.; Yang, Y. Pancreatic stellate cells derived exosomal miR-5703 promotes pancreatic cancer by downregulating CMTM4 and activating PI3K/Akt pathway. Cancer Lett. 2020, 490, 20–30. [Google Scholar] [CrossRef]
  88. Ma, Q.; Wu, H.; Xiao, Y.; Liang, Z.; Liu, T. Upregulation of exosomal microRNA-21 in pancreatic stellate cells promotes pancreatic cancer cell migration and enhances Ras/ERK pathway activity. Int. J. Oncol. 2020, 56, 1025–1033. [Google Scholar] [CrossRef] [PubMed]
  89. Cao, W.; Zeng, Z.; He, Z.; Lei, S. Hypoxic pancreatic stellate cell-derived exosomal mirnas promote proliferation and invasion of pancreatic cancer through the PTEN/AKT pathway. Aging 2021, 13, 7120–7132. [Google Scholar] [CrossRef] [PubMed]
  90. Perera, C.; Xu, Z.; Mekapogu, A.R.; Hosen, S.Z.; Pothula, S.; Greenfield, J.; Chari, S.; Goldstein, D.; Pirola, R.; Wilson, J.; et al. 1133 Pancreatic Stellate Cell and Cancer Cell Derived Exosomes Impair Beta Cell Function: Implications for Pancreatic Cancer Related Diabetes. Gastroenterology 2020, 158, S-221. [Google Scholar] [CrossRef]
  91. Valerio, A.; Basso, D.; Fogar, P.; Falconi, M.; Greco, E.; Bassi, C.; Seraglia, R.; Abu-Hilal, M.; Navaglia, F.; Zambon, C.-F.; et al. Maldi-TOF analysis of portal sera of pancreatic cancer patients: Identification of diabetogenic and antidiabetogenic peptides. Clin. Chim. Acta 2004, 343, 119–127. [Google Scholar] [CrossRef]
  92. Zhou, W.; Wang, Y.; Gao, H.; Jia, Y.; Xu, Y.; Wan, X.; Zhang, Z.; Yu, H.; Yan, S. Identification of Key Genes Involved in Pancreatic Ductal Adenocarcinoma with Diabetes Mellitus Based on Gene Expression Profiling Analysis. Pathol. Oncol. Res. 2021, 27, 604730. [Google Scholar] [CrossRef]
Figure 1. Biogenesis and the secretion of exosomes. Exosome biosynthesis begins during the maturation of the early sorting endosome into the late sorting endosome or multivesicular body (MVB). Exosome precursors known as intraluminal vesicles (ILVs) are contained within MVBs. MVBs can either be degraded (via fusion with lysosomes) or transported to the plasma membrane. Then, ILVs are released as exosomes into body fluids during the fusion of MVBs with the plasma membrane.
Figure 1. Biogenesis and the secretion of exosomes. Exosome biosynthesis begins during the maturation of the early sorting endosome into the late sorting endosome or multivesicular body (MVB). Exosome precursors known as intraluminal vesicles (ILVs) are contained within MVBs. MVBs can either be degraded (via fusion with lysosomes) or transported to the plasma membrane. Then, ILVs are released as exosomes into body fluids during the fusion of MVBs with the plasma membrane.
Ijms 24 10203 g001
Figure 2. Exosome release and uptake. Exosomes released by parent cells (tumour cells) can be taken up via the receptors on recipient cells or through mechanisms such as direct membrane fusion, phagocytosis, macropinocytosis and clathrin-dependent endocytosis. Exosomal cargo: green = proteins, yellow = lipids, grey = nucleic acids.
Figure 2. Exosome release and uptake. Exosomes released by parent cells (tumour cells) can be taken up via the receptors on recipient cells or through mechanisms such as direct membrane fusion, phagocytosis, macropinocytosis and clathrin-dependent endocytosis. Exosomal cargo: green = proteins, yellow = lipids, grey = nucleic acids.
Ijms 24 10203 g002
Figure 3. Integrated schematic workflow elucidating the potential mechanism of action of pancreatic cancer (PC)-derived adrenomedullin (AM) on PC cells and β cells. (a) AM may be present in tumour cell-derived exosomes either alone or (b) Together with CA 19-9, (c) Alternative channels for AM secretion are yet to be brought to scientific limelight. AM may then further act upon PC cells in an autocrine manner and on β cells in a paracrine manner with a variety of pro-tumorigenic and pro-diabetogenic effects, respectively. ? = unknown.
Figure 3. Integrated schematic workflow elucidating the potential mechanism of action of pancreatic cancer (PC)-derived adrenomedullin (AM) on PC cells and β cells. (a) AM may be present in tumour cell-derived exosomes either alone or (b) Together with CA 19-9, (c) Alternative channels for AM secretion are yet to be brought to scientific limelight. AM may then further act upon PC cells in an autocrine manner and on β cells in a paracrine manner with a variety of pro-tumorigenic and pro-diabetogenic effects, respectively. ? = unknown.
Ijms 24 10203 g003
Figure 4. Interactions of pancreatic stellate cells (PSCs) and tumour cells lead to the release of exosomes which could be taken up by (i). β cells resulting in decreased insulin secretion, and (ii). adipocytes, hepatocytes and/or myocytes, could result in impaired insulin signalling and insulin resistance.
Figure 4. Interactions of pancreatic stellate cells (PSCs) and tumour cells lead to the release of exosomes which could be taken up by (i). β cells resulting in decreased insulin secretion, and (ii). adipocytes, hepatocytes and/or myocytes, could result in impaired insulin signalling and insulin resistance.
Ijms 24 10203 g004
Table 1. Differences between PCRD and T2DM.
Table 1. Differences between PCRD and T2DM.
S/NPCRDT2DM
1.Patients develop DM despite preceding weight lossPatients gain weight at the time of DM onset
2.Occurs many months before the occurrence of cachexia (with greater than a 5% weight loss over 6 months)Not associated with cachexia
3.Occurs within 3–5 years before the clinical diagnosis of PCOccurs with or without PC development
4.Insulin levels are normal or lowInsulin levels are high and there is a marked insulin resistance
5.Usually improves or is resolved following PC treatments, including the surgical resection of the tumourPersistent DM accompanies PDAC patients with long-standing T2DM even after surgical resection
6.Low levels of the glucose-dependent insulinotropic polypeptide (GIP) are evidentVariable levels of GIP
7.Pancreatic polypeptide (PP) levels are low or absentPP levels are high or normal
8.Low glucagon levelsHigh glucagon levels
9.Occurs at any ageOccurs mainly in adulthood
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Binang, H.B.; Perera, C.J.; Apte, M.V. Role of Pancreatic Tumour-Derived Exosomes and Their Cargo in Pancreatic Cancer-Related Diabetes. Int. J. Mol. Sci. 2023, 24, 10203. https://doi.org/10.3390/ijms241210203

AMA Style

Binang HB, Perera CJ, Apte MV. Role of Pancreatic Tumour-Derived Exosomes and Their Cargo in Pancreatic Cancer-Related Diabetes. International Journal of Molecular Sciences. 2023; 24(12):10203. https://doi.org/10.3390/ijms241210203

Chicago/Turabian Style

Binang, Helen B., Chamini J. Perera, and Minoti V. Apte. 2023. "Role of Pancreatic Tumour-Derived Exosomes and Their Cargo in Pancreatic Cancer-Related Diabetes" International Journal of Molecular Sciences 24, no. 12: 10203. https://doi.org/10.3390/ijms241210203

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop