TY - JOUR AU1 - Lewis, Phillip L. AU2 - Wells, James M. AB - Abstract Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand β-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and β-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human β-cells, to cell lines, to pluripotent stem cell-derived β-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are β-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying β-cell biology, improving β-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic. Engineered in vitro systems have the potential to revolutionize our understanding metabolic diseases and the countless roles β-cells play in them. Many different sources of insulin-secreting cells are available for engineered systems each with their own strengths and weaknesses pertinent specific downstream applications. For example, high throughput studies require large numbers of cells allowing cell lines and stem cell-derived β-cells to be suitable sources. However, functional studies into β-cell physiology as well as organ-organ interactions require physiologically mature cell types that more closely mimic human biology. This review covers specific sources insulin-secreting cells as well as applications in fluidic culture devices and high throughput methodologies as they pertain to the study of β-cell physiology with a focus on diabetes. Open in new tabDownload slide Open in new tabDownload slide cell culture, diabetes, experimental models, pancreas, pancreatic differentiation, pluripotent stem cells, technology Significance statement While superficially a disease of insulin secretion and sugar metabolism, the deeper complexities of diabetes necessitate advanced engineered strategies to uncover new treatment approaches. In vitro models of β-cells and insulin secretion in a human-specific context have therefore emerged as powerful tools to study diabetes. This article reports several advances including different sources of insulin-secreting cells, organ-on-a-chip models, and high throughput strategies designed to deepen our understanding of β-cell biology and to better treat disease. INTRODUCTION Diabetes is a pervasive and growing problem across the world. With age-adjusted diabetes prevalence increasing from 4.3% in 1980 to nearly 9% in 2016 and over 415 million people worldwide living with diabetes,1,2 there is a clear need for the development of novel therapies to combat the loss and dysfunction of β-cells. In type 1 diabetes (T1D) where β-cells are absent, glycaemia can be effectively managed with exogenous insulin therapy. While managing glucose has been highly burdensome historically, new technologies such as continuous glucose monitoring (CGM) and insulin pumps have provided a much-improved quality of life. The majority of people with diabetes have type 2 diabetes (T2D) (90%-95%). T2D can be mitigated with lifestyle changes, or using pharmaceutical approaches that center around increasing peripheral tissue insulin sensitivity (eg, metformin), increasing insulin secretion from existing β-cells (eg, sulfonylureas), or by altering hormonal interaction with other endocrine organs (eg, incretin mimetics, DPP-IV inhibitors). By far the most successful treatment for T2D and its associated sequelae is gastric bypass surgery, which results in rapid restoration of β-cell function as well as improved liver and cardiovascular function before appreciable weight loss occurs.3,4 It is not known how gastrointestinal surgery results in a reversal of T2D but highlights the role that interorgan communication plays in healthy and diseased β-cell physiology. A deeper understanding of islet and β-cell physiology and how soluble factors from other organs feed into the restoration of normoglycemia is paramount to developing novel therapies for diabetes. Historically, animal models have provided the most insight into islet pathophysiology. Significant differences between humans and model organisms have led to drugs failing during clinical trials; for example, troglitazone was safe in animal testing but hepatotoxicity led to it being removed from American markets.5 The preponderance of examples of drugs passing preclinical and phase 1 trials but failing at later stages highlights the need for human cell and tissue model systems to study human physiology, diseases, toxicity and drug efficacy. The convergence of multiple fields ranging from developmental biology, endocrinology, chemistry, biomaterials science, and chemical engineering has brought forth numerous platforms to study many different aspects of islets and insulin-secreting cells. For example, physiologically responsive human β-like cells can now be derived from pluripotent stem cells (PSCs) and sophisticated engineered systems can precisely stimulate and measure islet and β-cell physiology. Moreover, it is now possible to generate other metabolically critical organ tissues in vitro and study their interactions with β-cells in efforts to model T2D. In this review, we will discuss recent progress in the development and use of human in vitro model systems to study β-cells, from cell sources to their applications in fluidic systems and in high throughput strategies. SOURCES OF INSULIN-PRODUCING CELLS The choice of a cell source for studying diabetes and islet or β-cell physiology ultimately depends on the application in question (Figure 1). Studying the role of a gene in islet development, for example, is better-suited for rodent or PSC-based approaches as opposed to cell lines or primary isolated human islets. However, human islets may be better-suited to study human-specific insulin secretion physiology given the relative immaturity of PSC-β-cells and differences between humans and model organisms. Each source has its own unique strengths and weaknesses in the context of in vitro studies, with some experimental questions necessitating more than one approach. FIGURE 1 Open in new tabDownload slide Sourcing of insulin producing cells for in vitro engineered systems. Primary isolated human islets are the closest approximation to what would be seen in vivo; however, drawbacks in variability are often a concern. Pseudoislets are an approach to mitigate some variability. PSC-derived sources offer a unique approach to study human physiology, while model organisms can place islet physiology in a whole-organism context. β-Cell lines are by far the easiest to use of all categories; however, they are perhaps the least physiologically representative of their in vivo counterparts FIGURE 1 Open in new tabDownload slide Sourcing of insulin producing cells for in vitro engineered systems. Primary isolated human islets are the closest approximation to what would be seen in vivo; however, drawbacks in variability are often a concern. Pseudoislets are an approach to mitigate some variability. PSC-derived sources offer a unique approach to study human physiology, while model organisms can place islet physiology in a whole-organism context. β-Cell lines are by far the easiest to use of all categories; however, they are perhaps the least physiologically representative of their in vivo counterparts Model organisms Model organisms have remained the gold standard for islet and diabetes research. Research findings around islet development and function have been remarkably reproducible due to the consistency of inbred animals. Furthermore, islet preparations for in vitro experiments are similar batch-to-batch and lab-to-lab. Cryopreserved rodent islets are also readily available for purchase from several sources. Nevertheless, there are structural and functional differences between human and rodent islets. Mouse and rat islets highly express the GLUT2 receptor, making them more susceptible to streptozotocin (STZ)-induced β-cell death and are widely used as in vivo model of islet destruction during diabetes.6 Mouse and human islets are structurally different, with mouse islets being half the size of human islets, with endocrine cells that are organized differently.7 Human islets also have a unique vasculature8 and undergo unique changes in response to disease compared with rodents.9 The functional differences in insulin secretion physiology between human and mouse β-cells have been thoroughly reviewed.10 In summary, human islets are sensitive to lower concentrations of glucose and the differences in oscillatory waves of calcium influx and insulin secretion may ultimately be due to architectural differences. However, due to the limited availability and batch to batch variability, rodents remain a powerful tool for mechanistic studies of islet development and disease both in vivo and in vitro. Aside from rodents, many other vertebrate models have been used for diabetes and islet research. Fish and frogs have been used to study pancreas development because of the ease of embryonic manipulation and in the case of zebrafish their genetic tractability and transparency.11,12 Larger animals such as nonhuman primates (NHP) and swine are not typically used in early-stage basic research because of ethical issues and cost. However, NHPs have been used as models for xenotransplantation and immunoisolation strategies13 in testing porcine islets as a possible therapeutic in humans.14 While there are functional differences in insulin secretion between porcine, NHP, and human islets,15 the availability of porcine islets is a considerable advantage of xenotransplantation over allotransplantation.16 Human Cadaveric human islets are a first choice for studying human physiology in vitro, and their availability in recent years has improved. However, these islets are often those rejected for transplantation and are of variable quality, resulting in significant differences in function between batches of islets.17 This variability can be mitigated with the formation of “pseudoislets,” which are enzymatically dissociated islets that are reaggregated into uniformly sized spheroids.17 While an aggregate of any insulin-producing cell, including cell lines can be termed a pseudoislet,18 for the purposes of this review, pseudoislets will refer to reaggregated primary islets. Pseudoislets are more uniform in size and in nutrient responsiveness, however, tend to have lower overall insulin content than their unmanipulated islet counterparts.17,19 The dissociation and reaggregation also allows for efficient viral transduction for manipulation of gene expression and introduction of live imaging reporters.20-22 Another configuration of islets is monolayer culture, which are more uniform and have been used for high throughput applications (see below). However, the dispersing of islet cells and growth on monolayers fundamentally changes islet architecture and thus is disruptive to insulin secretion physiology. The restored cell-cell contacts of pseudoislets rescues calcium physiology and allows for the study of interendocrine cell communication between β-cells and other islet cell types.23 Human islets have also been used therapeutically by transplantation into patients with difficult-to-control “brittle” T1D. Islet transplantation typically follows the Edmonton protocol where islets are enzymatically liberated from the rest of the pancreas and infused into the hepatic portal vein, allowing engraftment into hepatic lobules. During Total Pancreatectomy with Islet Autotransplantation (TPIAT) for patients with chronic pancreatitis, a patient's own islets are subjected to this procedure. While many institutions are developing methods and materials to improve survival and engraftment of transplanted islets, the insurmountable obstacle to the wide use of cadaveric islets for therapy is their limited availability. Furthermore, insulin dependence, along with other complications, may return in many patients receiving allogeneic islets due to many factors not limited to autoimmune destruction.24 For these reasons, the field of transplantation-based therapeutics has focused on a promising and unlimited source of insulin-expressing cells, human PSCs. PSC-derived The past decade has seen remarkable advances in the development of β-like cells and islets derived from PSCs as reviewed in.25,26 PSC-derived β-like cells can be generated in an unlimited and reproducible manner, are of consistent quality, and can be autologous for autotransplantation. Several companies (Sigilon/Eli Lilly,27 Evotec/Sanofi, Novo Nordisk, Semma,28 Sernova,29 Seraxis,30 and Viacyte31) have been developing therapeutics for the treatment of T1D that combine PSC-derived islets with encapsulation devices that afford some level of immunoprotection. As an alternative to immune protective devices some groups are developing genetically modified “universal donor” iPS lines that can evade the immune system and prevent the need for immunosuppressants. The goal is to develop an implantable and easily-retrievable biologic that will serve as a surrogate endocrine pancreas. While human PSCs have been transformative to study human β-cell development, most early protocols only were able to generate embryonic precursors that required transplantation and maturation in rodents to become nutrient responsive β-like cells.32 The more recent generation of protocols have been able to show nutrient responsiveness in static conditions28,33,34 and even dynamic glucose responses including first and second phase secretion profiles.35-37 While these cells are still not molecularly or functionally equivalent to human β-cells, there is every reason to believe that additional advances will continue to improve β-like cell function. For example, alternating 12 hours cycles of high and low glucose37 or cell sorting36 and assembly of β-cell-like aggregates35,38,39 enhances many aspects of β-like cell maturation such as mitochondria number/density and insulin secretion dynamics. Applications of PSC-based approaches to model diseases (including diabetes) have yielded a wealth of information on the roles genes play in islet differentiation and function.25 For example, increased susceptibility to gluco/lipotoxicity, both in vitro and in vivo was demonstrated using PSC-β-cells with mutated genes associated with T2D (CDKAL1, MT1E, KCNJ11, KCNQ1).40,41 However, T1D or T2D are rarely monogenic diseases, and in vitro modeling of T2D especially will require long-term culture of functionally mature β-like cells to adequately mimic disease progression. Cell lines The consistency and reproducibility of cell lines combined with their ease of use and maintenance make them indispensable tools for molecular biological studies. Many different insulin-secreting cell lines have been developed from human and rodent β-cells through SV40 immortalization or isolated insulinomas as reviewed in.42 Regardless of the source of immortalization, the primary disadvantage of β-cell lines is their lack of physiologic responsiveness to nutrients as evident in tests of glucose-stimulated insulin secretion (GSIS). β-Cell lines can be hypersensitive to glucose, with other lines insensitive to it, while some become progressively more insensitive as passage number increases. Although they differ from line to line, β-cell lines are generally less mature than human islets. Transciptomic and proteomic comparisons between β-cell lines and primary human islets confirm that there are many differences.43,44 The appropriate glucose responsiveness of the rat insulinoma cell line INS-1 and the mouse insulinoma cell line MIN6 (in the presence of nicotinamide) has made them a popular first choice in the development of engineered systems to study insulin secretion.45,46 Historically, few human β-cell lines have been available to study insulin secretion. The human β-cell EndoC-βH1 was developed by Ravassard et al by transplanting immortalized human fetal pancreatic bud tissue into immunocompromised mice.47 While more similar to human fetal β-cells, this line shows remarkable glucose responsiveness in vitro and upon transplantation in addition to not expressing markers of other pancreatic cell types. A similar cell line, EndoC-βH3, expresses CRE-ERT2, allowing its proliferation to be arrested upon exposure to tamoxifen.48 Cell lines whose proliferation is easily controllable (EndoC-βH3, R7T1) are excellent candidates for studies into β-cell proliferation. The insulin secretion efficacy of most cell lines can also be enhanced with cell aggregation.49-53 Therefore, β-cell line aggregates cultured in this manner serve as low-cost surrogates for islets/pseudoislets. ENGINEERED FLUIDIC PLATFORMS FOR STUDYING ISLETS AND INSULIN SECRETION PHYSIOLOGY The study of islet physiology and (dys)function with fluidic systems has emerged as an entire subfield of tissue-on-a-chip research due to the many advantages of flow systems in studying dynamic insulin secretion. By mimicking blood flow, β-(like)-cells are exposed to a nutrient concentration that is not impacted by the β-cells' own secretions and metabolism. Furthermore, secreted hormones are immediately swept away from β-cells, mitigating any autocrine effects of insulin that may not be physiologically relevant. Fluidic systems also have the advantage of constant sampling, allowing for a more detailed, time-resolved, resolution of an islet's secretion profile, and in some cases allowing for measurement of several different physiologic phenomena on a per-cell or per-islet basis. Fluidic devices have been well-suited to investigate the effects of fluid flow on islet function. Fluid flow enhances the viability, insulin secretion, and response to secretagogues of rat islets when compared with a static system54 and can improve GSIS of pseudoislets.55 Flow was also shown to lead to other morphological changes such as more abundant microvilli and increased amounts of tight junctions. The impact of flow on may impart beneficial effects on resident islet endothelial cells, indirectly improving islet function and viability.55,56 Numerous labs have devised platforms for analysis of islet secretions, which has been thoroughly reviewed previously.57-59 Discussed below are recent advances and new directions the organ-on-a-chip field has recently taken with β-cell research. Using fluidic systems to study β-cell physiology Fluidic systems for analysis of β-cell secretion, regardless of endpoint application, have settled on several common approaches. These include an islet and cell aggregate trap system which gently immobilizes cells and enables fluid contact with and flow around the islet surface. Islets can be trapped using a mesh system,60 with narrowing of fluid channels,61,62 or with troughs into which islets and cell aggregates will settle.63 Alternatively, pseudoislets can be formed on the chip itself which can then be perfused during long-term culture.55,64 Fluidic systems have also incorporated on-chip analysis or sample collection for subsequent analysis, reducing workload and enhancing the number of samples able to be collected compared with traditional methods (Figure 2). Commercially available fluidic platforms such as those manufactured by Biorep65 allow for sample collection, while Seahorse by Agilent66 systems continuously measure metabolic activity in a well plate format and allow for fluid collection for off-platform analysis. On-chip analysis dramatically accelerates workflow with the compromise of increased cost and occasionally reduced versatility in terms of output measurement options. On-chip readouts in through live imaging or electrodes, for example, do not require fluid sampling, and therefore enable the same off-chip quantifications to be performed without sacrificing sample volume. FIGURE 2 Open in new tabDownload slide Quantitatively measuring islet and β-cell physiology. Measurements from whole islets typically focus on peptide hormone secretion. Any method developed to quantify secreted peptides can therefore be used, with some having more utility for certain proteins (eg, insulin). Interrogating individual β-cells focuses on specific aspects of GSIS physiology, listed adjacent to each output are examples of detection methods. Influx/efflux of several ions can be quantified using fluorescent dyes or indicator proteins. Patch clamping can also be geared toward specific ions. Indicator proteins have also been used to study protein/vesicle trafficking as well as mitochondrial redox state. Many different dyes and fluorescent proteins have been developed to measure these β-cell functions. APG, Asante potassium green; ELISA, enzyme-linked immunosorbent assay; GEPIIs, genetically-encoded potassium ion indicators; GLP-1, glucagon-like peptide 1; IAPP, islet amyloid polypeptide; R123, rhodamine-123 FIGURE 2 Open in new tabDownload slide Quantitatively measuring islet and β-cell physiology. Measurements from whole islets typically focus on peptide hormone secretion. Any method developed to quantify secreted peptides can therefore be used, with some having more utility for certain proteins (eg, insulin). Interrogating individual β-cells focuses on specific aspects of GSIS physiology, listed adjacent to each output are examples of detection methods. Influx/efflux of several ions can be quantified using fluorescent dyes or indicator proteins. Patch clamping can also be geared toward specific ions. Indicator proteins have also been used to study protein/vesicle trafficking as well as mitochondrial redox state. Many different dyes and fluorescent proteins have been developed to measure these β-cell functions. APG, Asante potassium green; ELISA, enzyme-linked immunosorbent assay; GEPIIs, genetically-encoded potassium ion indicators; GLP-1, glucagon-like peptide 1; IAPP, islet amyloid polypeptide; R123, rhodamine-123 Some immediate changes in islets that occur upon exposure to a nutrient stimulus can only be observed with on-chip analysis. For example, upon exposure to glucose, β-cells will depolarize leading to an immediate influx of calcium ions which leads to vesicle fusion and insulin secretion. Controlled fluid exchange enables more precise exposure of cells to nutrients/secretagogues compared with manual exchanging of media. Continuous imaging correlated with fluid flow dramatically increases data fidelity by reducing error that may occur with timing or manual pipetting. Calcium flux can be imaged using indicator dyes such as Fura-2/4 am63 or Fluo-264 or using calcium-responsive proteins such as GCaMP6.22,67 Imaging platforms also enable a large number of islets to be simultaneously interrogated for calcium flux in response to hypoxia and secretagogues.68 The precision of engineered systems can exacerbate the inherent variability in biological systems, and incorporating large numbers of islets within a perfusion system can help smooth over that variability. β-Cells are highly metabolically active, with perturbations in mitochondrial activity being exceptionally relevant to diabetes.69 Dynamic mitochondrial activity can be visualized with Rhodamine 123.70 Glutathione redox state and hydrogen peroxide concentration of rat islet mitochondria were monitored real time in a perfusion system employing mitochondria-targeted ratiometric fluorescent proteins.71 Zbinden et al devised a system using Raman microspectroscopy to study multiple aspects of EndoC-βH31 aggregate function simultaneously in real time.72 Multireadout systems have also been designed to merge immunoassays and imaging,73 with some incorporating optogenetics to further interrogate islet function.74 Insulin secretion is by far the most popular islet output and can be measured in real time or in downstream off-chip analysis. Insulin secretion itself can be indicative of islet health if isolated from patients with diabetes.75 Continuous real-time on-chip measurements of insulin output can be taken using fluorescence anisotropy from islet arrays62 or single islets.76 This approach offers a uniquely precise and uninterrupted measurement rarely seen in perfusion platforms. Insulin secretion from a single pseudoislet was able to be quantified (off chip) using a hanging drop perifusion system.77 In single islet scenarios, higher resolution aspects of insulin secretion physiology can be observed such as oscillations during second phase insulin secretion which has previously only been shown with live Ca2+ imaging.78,79 Features such as the brief dip in first phase insulin secretion have never been shown in human islets. Furthermore, finer details in insulin secretion are essentially impossible to observe in static traditional culture methods. Continuous live monitoring or rapid sampling of small numbers of islets (or single islets) is therefore the optimal choice if such features of insulin secretion need to be observed. The time-resolution and range of concentrations of a flow system are the two factors that ultimately determine which insulin secretion detection method is most appropriate. Technical descriptions of different islet secretion detection methods are reviewed in.58 An indirect way to measure insulin secretion is using dyes that detect the zinc that is incorporated into insulin secretory vesicles.80-83 Potassium efflux, although an intermediate step in the progression of GSIS, has been measured within islets using fluorescence dyes.84 Schulze et al devised a platform with immediate measurements of NAD(P)H using fluorescence imaging, and oxygen consumption rate with O2 sensors.63 This was combined with buffer collection and subsequent off-chip insulin quantification. Glucagon output from PSC-derived cells has been measured using BioRep platforms33,36,37,85; however, other protein output that could provide important information about islet function (islet amyloid polypeptide [IAPP], somatostatin, pancreatic polypeptide [PP], ghrelin) are rarely measured. In one case a more sensitive technique, surface plasmon resonance (SPR), was used to quantify islet output of insulin, glucagon, and somatostatin.86 These multiparametric techniques allow for complex physiological studies of islet hormone secretions in response to nutrients and therapeutics. While in-depth analysis of sampled media is possible with off-chip techniques such as proteomics, continuous monitoring with methods such as imaging or SPR afford a time-resolved view of β-cell physiology. While there have been progress in simplifying perfusion systems,87 most of these technologies require expensive specialized equipment and skills that are not easily transferrable to other laboratories. Organs on a chip: The emergence of microphysiologic systems to study organ function and interaction As described above, fluidic systems have lent themselves to the development of microphysiological systems (MPSs) that allow for analysis of β-cell and islet function. The body of literature surrounding static coculture studies with islets and/or β-cells is extensive and largely suggest that coculture with nearly any cell type enhances β-cell function and viability.88,89 Alternatively, β-cell death can be stimulated by coculturing with immune cells in experiments designed to model immunological aspects of T1D. However, modeling functional interorgan crosstalk can be difficult to quantify in traditional static culture configurations. Engineered fluidic systems can enhance both control over multitissue interactions, as well as quantification of those interactions with many methods like those described in the previous section. MPSs have now been developed for most organ systems, including many organs that have endocrine interactions with the pancreas such as intestine, liver, adipose, muscle, and brain (Figure 3).90,91 Due to its essential role in drug metabolism, liver MPSs have been developed by several groups.92 Several platforms have been used to study liver function in the context of diabetes. Nonalcoholic fatty liver disease (NAFLD) is a diabetes comorbidity that has seen many engineered in vitro models.93-96 MPSs can be configured to study liver-pancreas communication and have been used to model how β-cells communicate to hepatocytes to store glycogen or release glucose in response to insulin and glucagon, respectively. Treating a fatty liver disease model with drugs like metformin, for example, would increase hepatocyte sensitivity to β-cell secreted insulin. In one example, pseudoislets impacted the activity of aggregates of the immortalized hepatocyte cell line HepaRG cells to monitor and take up glucose from the media.97 Moreover, coculture enhanced the function of both constructs after long-term (15 days) culture, indicating that hepatic secretions maintain pseudoislet function, and vice versa. This was also demonstrated with primary rat islets and hepatocytes.98 A β-cell/liver MPS could be used in the future to study pathologic triggers such as inflammatory cytokines and how they cause the β-cell and liver dysfunction that is associated with T2D.99 FIGURE 3 Open in new tabDownload slide Interorgan crosstalk with the endocrine pancreas. Summarized are influences each organ (system) has on pancreatic endocrine secretin. Listed beside each organ are the effects caused by hormones secreted by islets. While not an endocrine interaction, direct autonomic nervous system contact with islets influence endocrine secretion. The GI tract secretes ~20 separate hormones in addition to being responsible for nutrient absorption, shown here are hormones known to influence islet secretion. Three major energy storage/metabolism organs (liver, muscle, adipose) primarily influence pancreatic function by modulating blood glucose directly. However, they also modulate islet activity through endocrine interactions. ACh, acetylcholine; ACTH, adrenocorticotropic hormone; ADH, antidiuretic hormone—vasopressin; ANS, autonomic nervous system; BG, blood glucose; CNS, central nervous system; DPP4, dipeptidyl peptidase 4; EEC, enteroendocrine cell; FABP4, fatty acid-binding protein 4; FFA, free fatty acids; FGF21, fibroblast growth factor 21; GI, gastrointestinal; GIP, glucose insulinotropic polypeptide; GLP1/2, glucagon-like peptide 1, 2; NMU, neuromedin U; NPY, neuropeptide Y; PYY, peptide YY; SCFA, short chain fatty acids; SeP, selenoprotein P; SST, somatostatin; TG, triglycerides; TNF-α, tissue necrosis factor α; VIP, vasoactive inhibitory peptide FIGURE 3 Open in new tabDownload slide Interorgan crosstalk with the endocrine pancreas. Summarized are influences each organ (system) has on pancreatic endocrine secretin. Listed beside each organ are the effects caused by hormones secreted by islets. While not an endocrine interaction, direct autonomic nervous system contact with islets influence endocrine secretion. The GI tract secretes ~20 separate hormones in addition to being responsible for nutrient absorption, shown here are hormones known to influence islet secretion. Three major energy storage/metabolism organs (liver, muscle, adipose) primarily influence pancreatic function by modulating blood glucose directly. However, they also modulate islet activity through endocrine interactions. ACh, acetylcholine; ACTH, adrenocorticotropic hormone; ADH, antidiuretic hormone—vasopressin; ANS, autonomic nervous system; BG, blood glucose; CNS, central nervous system; DPP4, dipeptidyl peptidase 4; EEC, enteroendocrine cell; FABP4, fatty acid-binding protein 4; FFA, free fatty acids; FGF21, fibroblast growth factor 21; GI, gastrointestinal; GIP, glucose insulinotropic polypeptide; GLP1/2, glucagon-like peptide 1, 2; NMU, neuromedin U; NPY, neuropeptide Y; PYY, peptide YY; SCFA, short chain fatty acids; SeP, selenoprotein P; SST, somatostatin; TG, triglycerides; TNF-α, tissue necrosis factor α; VIP, vasoactive inhibitory peptide Other MPSs have been developed for tissues directly involved in the normal and pathologic handling of blood glucose including adipose tissue100,101 and skeletal muscle.102,103 A similar approach was used to study adipose tissue in engineered systems104 and combined islet/adipose systems.105 Insulin secretion was shown to be augmented in islets placed downstream of adipose tissue in an MPS.106 Studying interactions between β-cells and the major energy storage/metabolism organs (liver, adipose, muscle) is technically challenging in MPS. Culture media is typically used in volumes where any storage/release of glucose from these organs in response to insulin/glucagon would not be detectible. Other surrogate readouts for insulin stimulation (or lack thereof) such as presence of glycogen granules or AKT signaling can be used.107 Adipose-immune crosstalk has also been studied in the context of T2D in the absence of insulin-producing cells.108 The GI tract is collectively the largest endocrine organ in the human body and plays a central role in regulating the endocrine pancreas via its secreted hormones and absorbed macronutrients. Bariatric surgery itself is largely considered the only immediate “cure” for T2D, with an immediate cessation of symptoms observed before any loss of weight.109 This metabolic shift is largely thought to be due to the alterations in nutrient exposure to different regions of the gut, and the resulting changes in the secreted hormones.3 Hormones in the GI tract are secreted by enteroendocrine cells (EECs) which are exceedingly rare (1%-3% of the intestinal epithelium) making their study notoriously difficult. Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1), the “incretins,” are secreted by the EEC subtypes K and L cells, respectively, and are well-known to enhance glucose-stimulated insulin secretion. Several drugs are clinically available that harness the incretin effect.110,111 The L-cell line GLUTag was combined with INS-1 cells in an MPS to demonstrate an incretin effect, with dynamic secretion of both GLP-1 and insulin.112 However, as discussed above, the use of murine cells and transformed lines are not ideal models of human physiology. Moreover the intestine secretes >20 hormones whose influence on pancreatic endocrine hormone secretion is understudied,110 with other nonincretins such as PYY thought to also play a role in reversal of T2D postbariatric surgery.113 Therefore, developing primary human EEC MPSs to investigate the physiologic function and therapeutic potential of intestinal hormones in restoring β-cell function is paramount. Researchers have engineered platforms to explore vascularizing hydrogels containing encapsulated islets either in preparation for transplantation114-116 or for in vitro perfusion.56,117 Many of these in vitro systems are used in the study of T1D, often incorporating lymphocytes and vasculature into an engineered system to model autoimmune attack of islets.118 The destruction of vasculature due to diabetes was modeled independently of islets using PSC-derived endothelial cells.119 However, diabetes as a disease involves many tissues, necessitating design of more complex systems. The feasibility of a platform containing 4, 7, or 10 interconnected tissues (including islets) was demonstrated either driven by a pump120 or pumpless.121 These studies highlighted the compounding technical challenges that arise as each new MPS was added. Therefore, combining more than two tissues to effectively model endocrine disfunction in diabetes is a challenge, requiring a coordinated cross-disciplinary effort. Incorporating readouts in such complex systems will take the form of organ or cell-specific readouts (eg, Ca2+ indicator proteins) that are pertinent to a specific MPS design, or highly general approaches (eg, off-chip proteomics) to quantify tissue-tissue interactions. The widespread physiologic dysfunction of diabetes and the importance of insulin in everyday metabolism implies there is room for discovery in virtually any multitissue MPS involving islets.122-124 However, a pervasive shortcoming in the field is that new fluidic systems are not broadly adopted by other labs after the initial publication. It is common that there are no follow up publications, possibly due proprietary concerns because of attempts at commercialization, or the technical challenges of using these new complicated MPS systems. Also, the need to continue to develop new and better systems to publish papers is what pushes bioengineers to continuously design new platforms in lieu of using tools already at their disposal to probe deeper into β-cell physiology. These issues are being addressed by funding agencies, such as the NIH's National Center for Advancing Translational Sciences (NCATS) Tissue Chip program, which focuses on better sharing new MPS technologies with the broader research community. HIGH THROUGHPUT APPROACHES TO STUDY Β-(LIKE)-CELL BIOLOGY AND PHYSIOLOGY In order to be considered high throughput, a system must employ experimental replicates and dependent variables in quantities order(s) of magnitude higher than traditional approaches. This can be achieved through miniaturization, scale up, and automation of experimental techniques, data acquisition, and analysis. High throughput strategies involving islets and insulin-secreting cells focus primarily on identifying novel targets or drugs for the long-term goal of mitigating T1D or T2D. Historically, β cell lines have been the starting material of choice for HT screens due to the required scale up, minimal sample to sample variation, and two-dimensional (2D) configuration for imaging.125 However, technological advances such as robust protocols for PSC-derived β-like cells, rapid three-dimensional (3D) imaging, automated liquid handling, and automated image analysis have expanded researchers' capabilities to employ more physiologically mimetic systems in vitro.126 As discussed below many high-throughput screening (HTS) approaches focus on measuring changes in gene expression in 2D or 3D culture schemes, with detection of functional changes in HT occasionally requiring more nuance. 2D plate-based approaches 2D plate-based HT formats have been the gold standard of HT methods for decades. Superficially, cells are plated in a multiwell plate (96, 384, 1536-well plates), subjected to some treatment, and evaluated using automated image acquisition and analysis. These approaches have been applied to a wide variety of biological questions and are easily adaptable to evaluating islet and β-cell biology thanks to the availability of β-cell lines. One of the most evaluated aspects of β-cell biology in HT approaches is proliferation. Patients suffering from both T1D and T2D would benefit from enhanced β-cell proliferation; however, there are no clinically available drugs that are marketed for this purpose. Patients with both T1D and T2D may still have residual β-cells, which has led to a large research effort do identify compounds that can stimulate β-cell renewal.127 The R7T1 β-cell line contains a tet-on construct, only permitting its proliferation in the presence of a tetracycline. It is therefore an easy model to study β-cell proliferation in multiwell plates using viability-based assays (eg, CellTiter Glo).128,129 Images based assessment of cell proliferation via EDU incorporation and Ki67 staining has been used to identify novel genes involved in β-cell proliferation130 as well as compounds that enhance proliferation.131 Care should be taken using R7T1, as chemicals similar to tetracyclines may also induce proliferation.129 When dispersed primary islets are used, extra steps need to be taken to ensure proper physiological function and data integrity, such as pre-treating plates with extracellular matrix and omitting data from non-β-cells using image analysis, respectively.131 In that regard, an HT approach has also been developed to screen polymer surfaces for optimal attachment of dispersed islet cells, quantified with automated image analysis of insulin-expressing cells.132 2D plate-based HTS make excellent use of reporter-driven live cell automated imaging. Quantifying live changes in gene expression has unique utility in designing PSC differentiation protocols. Adapting PSCs to HT approaches has been thoroughly reviewed elsewhere.133 These approaches have helped define protocols that use small molecules that are now commonly used in many PSC differentiation protocols,134 including those designed to produce β-(like)-cells.135 The gradual replacement of recombinant proteins with small molecules is greatly reducing cost and encouraging more labs to explore stem cell differentiation protocols, with HTS playing an essential role. Reporter-driven HT approaches for β-cell differentiation center around increasing expression of genes critical to β-cell development, such as PDX1, NKX6.1, NEUROG3, MAFA and INS1, among others.136 Assaying for compounds that induce expression of certain transcription factors appropriate later in pancreatic differentiations, especially insulin, requires starting with cells already geared toward the pancreatic endoderm or endocrine precursors. For example HTS identified novel compounds that induce or enhance PDX1 expression during PSCs differentiation137,138 and in pancreatic ductal adenocarcinoma cell lines.139 Some approaches incorporate multiple gene reporters to screen for compounds that may enhance both PDX1 and INS1, for example.140 This same reporter system was used to identify a novel role of Na+ channels in modulating insulin expression.141 HTS was used to screen for the prohormone VGF,142 which has been shown to be essential in normal β-cell function and development.143 While cell lines or dispersed islets are well-suited in 2D HTS approaches, results are often validated using intact primary islets and animal models. The next generation of HTS could use physiological readouts such as insulin secretion or membrane depolarization, although adapting these to high throughput pose significant challenges, as described below. 3D approaches The frequent usage of imaging/image analysis in HT models poses unique challenges when employing more physiologically relevant 3D approaches. Advances in recent years in such as tissue clearing (eg, CLARITY144) and rapid 3D imaging platforms (eg, spinning disk, light sheet microscopy) have accelerated the merging of 3D models and HT technologies. A fluidic device combining on-chip culture, fixation, clearing, and imaging was demonstrated in a proof-of-concept study describing mouse islet vasculature in HT.145 Current costs of adopting these approaches has precluded widespread adoption, however. In that regard, simple epifluorescence plate-based imaging approaches can be applied to cell aggregates, such as dual INS1-NEUROG3 reporter transfected PSC aggregates.146 Higher resolution images of entire aggregates are not necessary in this specific application, as the presence of and ratios between two fluorescent reporters are the primary output. Automated image analysis can also be used for morphological analysis of islets encapsulated within biomaterials.147 This approach has been applied to encapsulation strategies148 and can be further applied to widely adopted immunoisolation strategies employed by several labs seeking to optimize islet health and engraftment upon transplantation,13,149,150 as well as stem cell differentiation.151 3D models have been applied where the primary output is not imaging-based, such as experiments studying secreted factors or lytic end point analyses. Aggregates of INS-1832/13 cells or human pseudoislets can be assayed for compounds that modulate static GSIS when placed in a 384-well plate.152 Aggregate-based approaches are often used in fluidic platforms to a certain HT degree, and may have several outputs quantified as discussed in the next section. Imaging aggregates in 3D has several technical challenges, such as movement of aggregates within the culture vessel because of plate scanning during image acquisition. Inspiration can be taken from approaches developed for whole organism HT studies using zebrafish. A workflow for HTS using transgenic insulin-YFP zebrafish that incorporates z-depth autofocusing and regional scanning/reporting in HT formats (termed automated reporter quantification in vivo [ARQiv]).153 This workflow was later applied to identify compounds that boost β-cell development154 or proliferation,155 taking advantage of the zebrafish's unique pancreas architecture. A separate group employed a fluorescent ubiquitylation-based cell cycle indicator (FUCCI) to identify compounds that induce proliferation in β-cells.156 Compounds that affect surrounding cells (islet vasculature, stroma) and indirectly enhance β-cell proliferation can be evaluated in whole-organisms HT scenarios. Other adverse effects, such as off-target carcinogenesis, can also be evaluated in this setting. HT functional studies High throughput approaches have enormous utility in probing β-cell biology. Understanding of the primary mechanisms behind β-cell apoptosis, metabolism, insulin secretion, and morphology can illuminate new opportunities for which therapeutics can be designed. However, designing HT studies based upon β-cell's physiological output can be technically challenging. Many of the studies above assay for changes in gene or protein expression; however, most of their follow-up validation studies that involve β-cell physiology do not qualify as HT. Indeed, reserving complex functional studies for low throughput validation is the primary reasoning for simplified, first-pass HT approaches. However, studying the biological end goal (eg, enhancing insulin secretion) in HT would potentially yield better results by streamlining workflow and cutting out intermediate steps. Several studies have measured static GSIS in HT in 2D arrays using cell lines,130,157-159 occasionally employing the use of automated liquid handling. Fluorescent dyes can also enable indirect HT static GSIS measurements via Ca2+ imaging using plate formats and automated liquid handling.129 However, PSC-based approaches have shown, Ca2+ imaging is not necessarily indicative of insulin secretion.160 As a method to employ fast and easy-to-use plate reader-based approaches, Min6 cells have been modified to cosecrete an insulin-luciferase fusion protein, and assayed for GSIS-enhancing compounds in HT.161 A similar technique was also used with INS1E cells157 to monitor GSIS. Other aspects of INS1E secretion physiology were quantified in scalable plate reader-based format using the membrane potential indicator dye DIBAC4(3), the Ca2+ dye Fura 2-AM, mitochondrial activity with rhodamine 123, and membrane lipid tension using FliptR.157 Several fluidic platforms can be used in HT; however, detecting secreted insulin in large volumes of fluid requires that several islets/aggregates be grouped together.64,65 The device designed by Lee et al involves 110 miniature bioreactors to study 1.1B4 cell line physiology under glucotoxic or lipotoxic conditions.64 This platform enables the formation of aggregates, live imaging, perfusion media sampling for GSIS, and cell lysis for genomic analysis all on the same culture device all in HT numbers. HT analysis of multiple biological processes of single islets using Raman microspectroscopy in a nondestructive manner was demonstrated in.72 Nondestructive techniques have potential applications in screening islets prior to transplantation without having to sacrifice tissue as scarce as islets. β-Cell death is involved in both T1D and T2D, and may be caused by immune-mediated destruction or exhaustion and gluco/lipotoxic conditions. Preventing β-cell destruction by these mechanisms is therefore a goal of several HT approaches that screen compounds that mitigate endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) as well apoptosis. As these processes are not exclusive to β-cells, several commercially available kits are for a variety of cellular processes can be easily scaled to HT levels. Some studies described in this section scaled popular MTT/XTT viability kits to HT.157,158,162 Dispersed human islets were used in a 384-plate-based siRNA screen, using automated liquid handling, to screen for novel antiapoptotic genes. Apoptosis as well as cellular reducing power were quantified using commercially available kits. Plate-based caspase activity assays (eg, Caspase-Glo) facilitate apoptosis quantification in 2D formats, while ER stress can be measured with CHOP-luciferase reporters.159 ER stress can also be measured indirectly by quantifying the ratio of XBP1 spliced/unspliced, in addition to quantifying phosphorylated IRE1-α protein, both of which can be done in HT.158 CONCLUSION Engineered human β-cells, β-cell functional readouts and engineered systems that allow for high throughput drug screening are all examples engineered-inspired approaches to study β-cells. Many of these systems have been used to uncover aspects of islet biology and insulin secretion physiology impossible to observe in traditional static or low-throughput systems. Features of dynamic insulin secretion profiles have been discovered as a result of single islet engineered systems. Novel roles of support cells within islets, such as endothelial cells, have been uncovered thanks to fluidic systems. Scientists and engineers are only beginning to scratch the surface of modeling interorgan crosstalk using MPS. MPS studies hold promise in the development of drugs that harness interorgan crosstalk, such as incretin-modifying drugs that modulate intestine-pancreas communication for the treatment of T2D. Many researchers are even using MPS to study ways to mitigate immune system attack of β-cells through islet vasculature. With the massive scale up underway, progressively more engineered strategies qualify as HT. HT strategies are already causing massive changes in the ways researchers evaluate efficacy of small molecule-induced changes in gene expression. New approaches to quantify β-cell function in HT can lead to the development of novel drugs, or drug classes, that can directly influence β-cell physiology. Moreover, researchers are developing novel ways to evaluate and enhance the physiology if engineered β-cells. Researchers have even created fully synthetic insulin secreting cells capable of responding to glucose.163 Furthermore, only briefly touched upon in this review are the remarkable advances achieved in engineered extracellular matrix platforms for islet culture and transplantation.164,165 The increased sensitivity, sophistication, and versatility of the engineered models described above have achieved profound impact in their ability further dissect islet biology and insulin secretion physiology. Future approaches combining many of the approaches outlined above will no doubt lead to significant discoveries eventually leading to vast improvements in the treatment of diabetes. ACKNOWLEDGMENTS J.M.W. and P.L.L. are supported by the NIH grants P01 HD093363 and UG3 DK119982 (J.M.W.), a Shipley Foundation award (J.M.W.), and an award from the Allen Foundation (J.M.W.). We would also like to thank Dr Heather A. McCauley for critical reading of this manuscript. AUTHOR CONTRIBUTIONS P.L.L.: manuscript conception, writing, editing, final approval; J.M.W.: manuscript conception, editing, final approval. CONFLICT OF INTEREST J.M.W. declared research funding from the NIH. P.L.L. declared no potential conflicts of interest. DATA AVAILABILITY STATEMENT Data sharing is not applicable to this article as no new data were created or analyzed in this study. REFERENCES 1 Collaboration NRF . Worldwide trends in diabetes since 1980: a pooled analysis of 751 population-based studies with 4.4 million participants . Lancet (London, England) . 2016 ; 387 : 1513 - 1530 . Crossref Search ADS PubMed WorldCat 2 Harding JL , Pavkov ME, Magliano DJ, Shaw JE, Gregg EW. Global trends in diabetes complications: a review of current evidence . Diabetologia . 2019 ; 62 : 3 - 16 . Google Scholar Crossref Search ADS PubMed WorldCat 3 Holst JJ . Enteroendocrine secretion of gut hormones in diabetes, obesity and after bariatric surgery . Curr Opin Pharmacol . 2013 ; 13 : 983 - 988 . Google Scholar Crossref Search ADS PubMed WorldCat 4 Gribble FM , Reimann F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism . Nat Rev Endocrinol . 2019 ; 15 : 226 - 237 . Google Scholar Crossref Search ADS PubMed WorldCat 5 Murphy EJ , Davern TJ, Shakil AO, et al. Troglitazone-induced fulminant hepatic failure . Dig Dis Sci . 2000 ; 45 : 549 - 553 . Google Scholar Crossref Search ADS PubMed WorldCat 6 Furman BL . Streptozotocin-induced diabetic models in mice and rats . Curr Protoc Pharmacol . 2015 ; 70 : 5.47.1 - 5.47.20 . Google Scholar Crossref Search ADS WorldCat 7 Dolenšek J , Rupnik MS, Stožer A. Structural similarities and differences between the human and the mouse pancreas . Islets . 2015 ; 7 : e1024405 . Google Scholar Crossref Search ADS PubMed WorldCat 8 Brissova M , Shostak A, Fligner CL, et al. Human islets have fewer blood vessels than mouse islets and the density of islet vascular structures is increased in type 2 diabetes . J Histochem Cytochem . 2015 ; 63 : 637 - 645 . Google Scholar Crossref Search ADS PubMed WorldCat 9 Kim A , Miller K, Jo J, Kilimnik G, Wojcik P, Hara M. Islet architecture: a comparative study . Islets . 2009 ; 1 : 129 - 136 . Google Scholar Crossref Search ADS PubMed WorldCat 10 Skelin Klemen M , Dolenšek J, Slak Rupnik M, Stožer A. The triggering pathway to insulin secretion: functional similarities and differences between the human and the mouse β cells and their translational relevance . Islets . 2017 ; 9 : 109 - 139 . Google Scholar Crossref Search ADS PubMed WorldCat 11 Kinkel MD , Prince VE. On the diabetic menu: zebrafish as a model for pancreas development and function . Bioessays . 2009 ; 31 : 139 - 152 . Google Scholar Crossref Search ADS PubMed WorldCat 12 Wallace KN , Pack M. Unique and conserved aspects of gut development in zebrafish . Dev Biol . 2003 ; 255 : 12 - 29 . Google Scholar Crossref Search ADS PubMed WorldCat 13 Bochenek MA , Veiseh O, Vegas AJ, et al. Alginate encapsulation as long-term immune protection of allogeneic pancreatic islet cells transplanted into the omental bursa of macaques . Nat Biomed Eng . 2018 ; 2 : 810 - 821 . Google Scholar Crossref Search ADS PubMed WorldCat 14 Cooper DKC , Ezzelarab MB, Hara H, et al. The pathobiology of pig-to-primate xenotransplantation: a historical review . Xenotransplantation . 2016 ; 23 : 83 - 105 . Google Scholar Crossref Search ADS PubMed WorldCat 15 Mueller KR , Balamurugan AN, Cline GW, et al. Differences in glucose-stimulated insulin secretion in vitro of islets from human, nonhuman primate, and porcine origin . Xenotransplantation . 2013 ; 20 : 75 - 81 . Google Scholar Crossref Search ADS PubMed WorldCat 16 Casu A , Bottino R, Balamurugan AN, et al. Metabolic aspects of pig-to-monkey (Macaca fascicularis) islet transplantation: implications for translation into clinical practice . Diabetologia . 2008 ; 51 : 120 - 129 . Google Scholar Crossref Search ADS PubMed WorldCat 17 Hilderink J , Spijker S, Carlotti F, et al. Controlled aggregation of primary human pancreatic islet cells leads to glucose-responsive pseudoislets comparable to native islets . J Cell Mol Med . 2015 ; 19 : 1836 - 1846 . Google Scholar Crossref Search ADS PubMed WorldCat 18 Persaud SJ , Arden C, Bergsten P, et al. Pseudoislets as primary islet replacements for research: report on a symposium at King's College London, London UK . Islets . 2010 ; 2 : 236 - 239 . Google Scholar Crossref Search ADS PubMed WorldCat 19 Yu Y , Gamble A, Pawlick R, et al. Bioengineered human pseudoislets form efficiently from donated tissue, compare favourably with native islets in vitro and restore normoglycaemia in mice . Diabetologia . 2018 ; 61 : 2016 - 2029 . Google Scholar Crossref Search ADS PubMed WorldCat 20 Harata M , Liu S, Promes JA, Burand AJ, Ankrum JA, Imai Y. Delivery of shRNA via lentivirus in human pseudoislets provides a model to test dynamic regulation of insulin secretion and gene function in human islets . Physiol Rep . 2018 ; 6 : e13907 . Google Scholar Crossref Search ADS PubMed WorldCat 21 Liu S , Harata M, Promes JA, et al. Lentiviral mediated gene silencing in human pseudoislet prepared in low attachment plates . J Vis Exp . 2019 ; 2019 : 1 - 9 . Google Scholar OpenURL Placeholder Text WorldCat 22 Walker JT , Haliyur R, Nelson HA, et al. Integrated human pseudoislet system and microfluidic platform demonstrate differences in GPCR signaling in islet cells . JCI Insight . 2020 ; 5 : 1 - 15 . Google Scholar Crossref Search ADS WorldCat 23 Reissaus CA , Piston DW. Reestablishment of glucose inhibition of glucagon secretion in small pseudoislets . Diabetes . 2017 ; 66 : 960 - 969 . Google Scholar Crossref Search ADS PubMed WorldCat 24 Nakamura T , Fujikura J, Anazawa T, et al. Long-term outcome of islet transplantation on insulin-dependent diabetes mellitus: an observational cohort study . J Diabetes Investig . 2020 ; 11 : 363 - 372 . Google Scholar Crossref Search ADS PubMed WorldCat 25 Balboa D , Saarimäki-Vire J, Otonkoski T. Concise review: human pluripotent stem cells for the modeling of pancreatic β-cell pathology . Stem Cells . 2019 ; 37 : 33 - 41 . Google Scholar Crossref Search ADS PubMed WorldCat 26 Amirruddin NS , Low BSJ, Lee KO, Tai ES, Teo AKK. New insights into human beta cell biology using human pluripotent stem cells . Semin Cell Dev Biol . 2020 ; 103 : 31 - 40 . Google Scholar Crossref Search ADS PubMed WorldCat 27 Bose S , Volpatti LR, Thiono D, et al. A retrievable implant for the long-term encapsulation and survival of therapeutic xenogeneic cells . Nat Biomed Eng . 2020 ; 4 : 814 - 826 . Google Scholar Crossref Search ADS PubMed WorldCat 28 Pagliuca FW , Millman JR, Gürtler M, et al. Generation of functional human pancreatic β cells in vitro . Cell . 2014 ; 159 : 428 - 439 . Google Scholar Crossref Search ADS PubMed WorldCat 29 Pepper AR , Pawlick R, Gala-Lopez B, et al. Diabetes is reversed in a murine model by marginal mass syngeneic islet transplantation using a subcutaneous cell pouch device . Transplantation . 2015 ; 99 : 2294 - 2300 . Google Scholar Crossref Search ADS PubMed WorldCat 30 Southard SM , Kotipatruni RP, Rust WL. Generation and selection of pluripotent stem cells for robust differentiation to insulin-secreting cells capable of reversing diabetes in rodents . PLoS One . 2018 ; 13 : 1 - 21 . Google Scholar Crossref Search ADS WorldCat 31 Agulnick AD , Ambruzs DM, Moorman MA, et al. Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo . Stem Cells Translational Medicine . 2015 ; 4 : 1214 - 1222 . Google Scholar Crossref Search ADS PubMed WorldCat 32 Schliesser JVWJ . Generation of beta cells from pluripotent stem cells: are we there yet? Ann N Y Acad Sci . 2014 ; 1311 : 124 - 137 . Google Scholar Crossref Search ADS PubMed WorldCat 33 Rezania A , Bruin JE, Arora P, et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells . Nat Biotechnol . 2014 ; 32 : 1121 - 1133 . Google Scholar Crossref Search ADS PubMed WorldCat 34 Russ HA , Parent AV, Ringler JJ, et al. Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro . EMBO J . 2015 ; 34 : 1759 - 1772 . Google Scholar Crossref Search ADS PubMed WorldCat 35 Velazco-Cruz L , Song J, Maxwell KG, et al. Acquisition of dynamic function in human stem cell-derived β cells . Stem Cell Rep . 2019 ; 12 : 351 - 365 . Google Scholar Crossref Search ADS WorldCat 36 Nair GG , Liu JS, Russ HA, et al. Recapitulating endocrine cell clustering in culture promotes maturation of human stem-cell-derived β cells . Nat Cell Biol . 2019 ; 21 : 263 - 274 . Google Scholar Crossref Search ADS PubMed WorldCat 37 Alvarez-dominguez JR , Donaghey J, Rasouli N, et al. Circadian entrainment triggers maturation of human in vitro islets resource circadian entrainment triggers maturation of human in vitro islets . Cell Stem Cell . 2020 ; 26 ( 1 ): 108 -122.e10. Google Scholar Crossref Search ADS PubMed WorldCat 38 Dayem AA , Bin LS, Kim K, et al. Recent advances in organoid culture for insulin production and diabetes therapy: methods and challenges . BMB Rep . 2019 ; 52 : 295 - 303 . Google Scholar Crossref Search ADS PubMed WorldCat 39 Tran R , Moraes C, Hoesli CA. Controlled clustering enhances PDX1 and NKX6.1 expression in pancreatic endoderm cells derived from pluripotent stem cells . Sci Rep . 2020 ; 10 : 1 - 12 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 40 Zeng H , Guo M, Zhou T, et al. An isogenic human ESC platform for functional evaluation of genome-wide-association-study-identified diabetes genes and drug discovery . Cell Stem Cell . 2016 ; 19 : 326 - 340 . Google Scholar Crossref Search ADS PubMed WorldCat 41 Guo M , Zhang T, Dong X, et al. Using hESCs to probe the interaction of the diabetes-associated genes CDKAL1 and MT1E . Cell Rep . 2017 ; 19 : 1512 - 1521 . Google Scholar Crossref Search ADS PubMed WorldCat 42 Skelin M , Rupnik M, Cencic A. Pancreatic beta cell lines and their applications in diabetes mellitus research . ALTEX . 2010 ; 27 : 105 - 113 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 43 Kutlu B , Burdick D, Baxter D, et al. Detailed transcriptome atlas of the pancreatic beta cell . BMC Med Genomics . 2009 ; 2 : 1 - 11 . Google Scholar Crossref Search ADS PubMed WorldCat 44 Ku GM , Kim H, Vaughn IW, et al. Research resource: RNA-seq reveals unique features of the pancreatic β-cell transcriptome . Mol Endocrinol . 2012 ; 26 : 1783 - 1792 . Google Scholar Crossref Search ADS PubMed WorldCat 45 Ishihara H , Asano T, Tsukuda K, et al. Pancreatic beta cell line MIN6 exhibits characteristics of glucose metabolism and glucose-stimulated insulin secretion similar to those of normal islets . Diabetologia . 1993 ; 36 : 1139 - 1145 . Google Scholar Crossref Search ADS PubMed WorldCat 46 Asfari M , Janjic D, Meda P, Li G, Halban PA, Wollheim CB. Establishment of 2-mercaptoethanol-dependent differentiated insulin-secreting cell lines . Endocrinology . 1992 ; 130 : 167 - 178 . Google Scholar Crossref Search ADS PubMed WorldCat 47 Ravassard P , Czernichow P, Ravassard P, et al. A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion . J Clin Investig . 2011 ; 121 : 3589 - 3597 . Google Scholar Crossref Search ADS PubMed WorldCat 48 Benazra M , Lecomte MJ, Colace C, et al. A human beta cell line with drug inducible excision of immortalizing transgenes . Mol Metab . 2015 ; 4 : 916 - 925 . Google Scholar Crossref Search ADS PubMed WorldCat 49 Tsonkova VG , Sand FW, Wolf XA, et al. The EndoC-βH1 cell line is a valid model of human beta cells and applicable for screenings to identify novel drug target candidates . Mol Metab . 2018 ; 8 : 144 - 157 . Google Scholar Crossref Search ADS PubMed WorldCat 50 Chowdhury A , Dyachok O, Tengholm A, Sandler S, Bergsten P. Functional differences between aggregated and dispersed insulin-producing cells . Diabetologia . 2013 ; 56 : 1557 - 1568 . Google Scholar Crossref Search ADS PubMed WorldCat 51 Bernard AB , Lin CC, Anseth KS. A microwell cell culture platform for the aggregation of pancreatic β-cells . Tissue Eng Part C Methods . 2012 ; 18 : 583 - 592 . Google Scholar Crossref Search ADS PubMed WorldCat 52 Nibbelink MG , Marchioli G, Moroni L, et al. A protocol to enhance INS1E and MIN6 functionality—the use of theophylline . Int J Mol Sci . 2016 ; 17 : 1 - 9 . Google Scholar OpenURL Placeholder Text WorldCat 53 Kuehn C , Dubiel EA, Sabra G, Vermette P. Culturing INS-1 cells on CDPGYIGSR-, RGD- and fibronectin surfaces improves insulin secretion and cell proliferation . Acta Biomater . 2012 ; 8 : 619 - 626 . Google Scholar Crossref Search ADS PubMed WorldCat 54 Essaouiba A , Okitsu T, Jellali R, et al. Microwell-based pancreas-on-chip model enhances genes expression and functionality of rat islets of Langerhans . Mol Cell Endocrinol . 2020 ; 514 : 110892 . Google Scholar Crossref Search ADS PubMed WorldCat 55 Jun Y , Lee JS, Choi S, et al. In vivo–mimicking microfluidic perfusion culture of pancreatic islet spheroids . Sci Adv . 2019 ; 5 : 1 - 13 . Google Scholar OpenURL Placeholder Text WorldCat 56 Sankar KS , Green BJ, Crocker AR, et al. Culturing pancreatic islets in microfluidic flow enhances morphology of the associated endothelial cells . PLoS One . 2011 ; 6 : 1 - 11 . Google Scholar Crossref Search ADS WorldCat 57 Sokolowska P , Janikiewicz J, Jastrzebska E, Brzozka Z, Dobrzyn A. Combinations of regenerative medicine and lab-on-a-chip systems: new hope to restoring the proper function of pancreatic islets in diabetes . Biosens Bioelectron . 2020 ; 167 : 112451 . Google Scholar Crossref Search ADS PubMed WorldCat 58 Castiello FR , Heileman K, Tabrizian M. Microfluidic perfusion systems for secretion fingerprint analysis of pancreatic islets: applications, challenges and opportunities . Lab Chip . 2016 ; 16 : 409 - 431 . Google Scholar Crossref Search ADS PubMed WorldCat 59 Wang Y , Lo JF, Mendoza-elias JE, et al. Application of microfluidic technology to pancreatic islet research: first decade of endeavor . Bioanalysis . 2010 ; 2 : 1729 - 1744 . Google Scholar Crossref Search ADS PubMed WorldCat 60 Hori T , Yamane K, Anazawa T, et al. Compact fluidic system for functional assessment of pancreatic islets . Biomed Microdevices . 2019 ; 21 : 1 - 9 . Google Scholar Crossref Search ADS WorldCat 61 Brooks JC , Ford KI, Holder DH, Holtan MD, Easley CJ. Macro-to-micro interfacing to microfluidic channels using 3D-printed templates: application to time-resolved secretion sampling of endocrine tissue . Analyst . 2016 ; 141 : 5714 - 5721 . Google Scholar Crossref Search ADS PubMed WorldCat 62 Glieberman AL , Pope BD, Zimmerman JF, et al. Synchronized stimulation and continuous insulin sensing in a microfluidic human islet on a chip designed for scalable manufacturing . Lab Chip . 2019 ; 19 : 2993 - 3010 . Google Scholar Crossref Search ADS PubMed WorldCat 63 Schulze T , Mattern K, Früh E, et al. A 3D microfluidic perfusion system made from glass for multiparametric analysis of stimulus-secretioncoupling in pancreatic islets . Biomed Microdevices . 2017 ; 19 : 1 - 11 . Google Scholar Crossref Search ADS PubMed WorldCat 64 Lee SH , Hong SG, Song J, et al. Microphysiological analysis platform of pancreatic islet β-cell spheroids . Adv Healthc Mater . 2018 ; 7 : 1 - 11 . Google Scholar OpenURL Placeholder Text WorldCat 65 Cabrera O , Jacques-Silva MC, Berman DM, et al. Automated, high-throughput assays for evaluation of human pancreatic islet function . Cell Transplant . 2008 ; 16 : 1039 - 1048 . Google Scholar Crossref Search ADS PubMed WorldCat 66 Yépez VA , Kremer LS, Iuso A, et al. OCR-stats: robust estimation and statistical testing of mitochondrial respiration activities using seahorse xf analyzer . PLoS One . 2018 ; 13 : 1 - 18 . Google Scholar Crossref Search ADS WorldCat 67 Pérez Koldenkova V , Nagai T. Genetically encoded Ca2+ indicators: properties and evaluation . Biochim Biophys Acta—Mol Cell Res . 2013 ; 1833 : 1787 - 1797 . Google Scholar Crossref Search ADS WorldCat 68 Nourmohammadzadeh M , Xing Y, Lee JW, et al. A microfluidic array for real-time live-cell imaging of human and rodent pancreatic islets . Lab Chip . 2016 ; 16 : 1466 - 1472 . Google Scholar Crossref Search ADS PubMed WorldCat 69 Martin SD , McGee SL. The role of mitochondria in the aetiology of insulin resistance and type 2 diabetes . Biochim Biophys Acta—Gen Subj . 2014 ; 1840 : 1303 - 1312 . Google Scholar Crossref Search ADS WorldCat 70 Lee D , Wang Y, Mendoza-Elias JE, et al. Dual microfluidic Perifusion networks for concurrent islet perifusion and optical imaging . Biomed Microdevices . 2012 ; 14 : 7 - 16 . Google Scholar Crossref Search ADS PubMed WorldCat 71 Roma LP , Duprez J, Takahashi HK, Gilon P, Wiederkehr A, Jonas JC. Dynamic measurements of mitochondrial hydrogen peroxide concentration and glutathione redox state in rat pancreatic β-cells using ratiometric fluorescent proteins: confounding effects of pH with HyPer but not roGFP1 . Biochem J . 2012 ; 441 : 971 - 978 . Google Scholar Crossref Search ADS PubMed WorldCat 72 Zbinden A , Marzi J, Schlünder K, et al. Non-invasive marker-independent high content analysis of a microphysiological human pancreas-on-a-chip model . Matrix Biol . 2020 ; 85–86 : 205 - 220 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 73 Yi L , Bandak B, Wang X, Bertram R, Roper MG. Dual detection system for simultaneous measurement of intracellular fluorescent markers and cellular secretion . Anal Chem . 2016 ; 88 : 10368 - 10373 . Google Scholar Crossref Search ADS PubMed WorldCat 74 Lenguito G , Chaimov D, Weitz JR, et al. Resealable, optically accessible, PDMS-free fluidic platform for ex vivo interrogation of pancreatic islets . Lab Chip . 2017 ; 17 : 772 - 781 . Google Scholar Crossref Search ADS PubMed WorldCat 75 Gomez Y , Navarro-Tableros V, Tetta C, et al. A versatile model of microfluidic perifusion system for the evaluation of C-peptide secretion profiles: comparison between human pancreatic islets and HLSC-derived islet-like structures . Biomedicine . 2020 ; 8 : 1 - 13 . Google Scholar OpenURL Placeholder Text WorldCat 76 Schrell AM , Mukhitov N, Yi L, Adablah JE, Menezes J, Roper MG. Online fluorescence anisotropy immunoassay for monitoring insulin secretion from islets of langerhans . Anal Methods . 2017 ; 9 : 38 - 45 . Google Scholar Crossref Search ADS PubMed WorldCat 77 Misun PM , Yesildag B, Forschler F, et al. In vitro platform for studying human insulin release dynamics of single pancreatic islet microtissues at high resolution . Adv Biosyst . 2020 ; 4 : 1 - 14 . Google Scholar Crossref Search ADS WorldCat 78 Bandak B , Yi L, Roper MG. Microfluidic-enabled quantitative measurements of insulin release dynamics from single islets of Langerhans in response to 5-palmitic acid hydroxy stearic acid . Lab Chip . 2018 ; 18 : 2873 - 2882 . Google Scholar Crossref Search ADS PubMed WorldCat 79 Li X , Hu J, Easley CJ. Automated microfluidic droplet sampling with integrated, mix-and-read immunoassays to resolve endocrine tissue secretion dynamics . Lab Chip . 2018 ; 18 : 2926 - 2935 . Google Scholar Crossref Search ADS PubMed WorldCat 80 Easley CJ , Rocheleau JV, Head WS, Piston DW. Quantitative measurement of zinc secretion from pancreatic islets with high temporal resolution using droplet-based microfluidics . Anal Chem . 2009 ; 81 : 9086 - 9095 . Google Scholar Crossref Search ADS PubMed WorldCat 81 Li D , Chen S, Bellomo EA, et al. Imaging dynamic insulin release using a fluorescent zinc indicator for monitoring induced exocytotic release (ZIMIR) . Proc Natl Acad Sci USA . 2011 ; 108 : 21063 - 21068 . Google Scholar Crossref Search ADS PubMed WorldCat 82 Radford RJ , Chyan W, Lippard SJ. Peptide-based targeting of fluorescent zinc sensors to the plasma membrane of live cells . Chem Sci . 2013 ; 4 : 3080 - 3084 . Google Scholar Crossref Search ADS PubMed WorldCat 83 Chen Y , Bai Y, Han Z, He W, Guo Z. Photoluminescence imaging of Zn2+ in living systems . Chem Soc Rev . 2015 ; 44 : 4517 - 4546 . Google Scholar Crossref Search ADS PubMed WorldCat 84 Bischof H , Rehberg M, Stryeck S, et al. Novel genetically encoded fluorescent probes enable real-time detection of potassium in vitro and in vivo . Nat Commun . 2017 ; 8 : 1 - 11 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 85 Zhu Y , Tonne JM, Liu Q, et al. Targeted derivation of organotypic glucose- and GLP-1-responsive β cells prior to transplantation into diabetic recipients . Stem Cell Rep . 2019 ; 13 : 307 - 321 . Google Scholar Crossref Search ADS WorldCat 86 Castiello FR , Tabrizian M. Multiplex surface plasmon resonance imaging-based biosensor for human pancreatic islets hormones quantification . Anal Chem . 2018 ; 90 : 3132 - 3139 . Google Scholar Crossref Search ADS PubMed WorldCat 87 Xing Y , Nourmohammadzadeh M, Elias JEM, et al. A pumpless microfluidic device driven by surface tension for pancreatic islet analysis . Biomed Microdevices . 2016 ; 18 : 80 . Google Scholar Crossref Search ADS PubMed WorldCat 88 de Souza BM , Bouças AP, de dos Oliveira FS, et al. Effect of co-culture of mesenchymal stem/stromal cells with pancreatic islets on viability and function outcomes: a systematic review and meta-analysis . Islets . 2017 ; 9 : 30 - 42 . Google Scholar Crossref Search ADS PubMed WorldCat 89 Lemos NE , de Almeida BL, Dieter C, et al. Use of additives, scaffolds and extracellular matrix components for improvement of human pancreatic islet outcomes in vitro: a systematic review . Islets . 2017 ; 9 : 73 - 86 . Google Scholar Crossref Search ADS PubMed WorldCat 90 Ronaldson-Bouchard K , Vunjak-Novakovic G. Organs-on-a-chip: a fast track for engineered human tissues in drug development . Cell Stem Cell . 2018 ; 22 : 310 - 324 . Google Scholar Crossref Search ADS PubMed WorldCat 91 Cui P , Wang S. Application of microfluidic chip technology in pharmaceutical analysis: a review . J Pharm Anal . 2019 ; 9 : 238 - 247 . Google Scholar Crossref Search ADS PubMed WorldCat 92 Deng J , Wei W, Chen Z, et al. Engineered liver-on-a-chip platform to mimic liver functions and its biomedical applications: a review . Micromachines . 2019 ; 10 : 1 - 26 . Google Scholar OpenURL Placeholder Text WorldCat 93 Gori M , Simonelli MC, Giannitelli SM, et al. Investigating nonalcoholic fatty liver disease in a liver-on-a-chip microfluidic device . PLoS One . 2016 ; 11 : 1 - 15 . Google Scholar OpenURL Placeholder Text WorldCat 94 Ouchi R , Togo S, Kimura M, et al. Modeling steatohepatitis in humans with pluripotent stem cell-derived Organoids . Cell Metab . 2019 ; 30 : 374 - 384.e6 . Google Scholar Crossref Search ADS PubMed WorldCat 95 da Silva Morais A , Oliveira JM, Reis RL. Biomaterials and microfluidics for liver models . Adv Exp Med Biol . 2020 ; 1230 : 65 - 86 . Google Scholar Crossref Search ADS PubMed WorldCat 96 Wang Y , Wang H, Deng P, et al. Modeling human nonalcoholic fatty liver disease (NAFLD) with an organoids-on-a-chip system . ACS Biomater Sci Eng . 2020 ; 6 ( 10 ): 5734 -5743. Google Scholar Crossref Search ADS PubMed WorldCat 97 Bauer S , Wennberg Huldt C, Kanebratt KP, et al. Functional coupling of human pancreatic islets and liver spheroids on-a-chip: towards a novel human ex vivo type 2 diabetes model . Sci Rep . 2017 ; 7 : 1 - 11 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 98 Essaouiba A , Okitsu T, Kinoshita R, et al. Development of a pancreas-liver organ-on-chip coculture model for organ-to-organ interaction studies . Biochem Eng J . 2020 ; 164 : 107783 . Google Scholar Crossref Search ADS WorldCat 99 Watt MJ , Miotto PM, De Nardo W, et al. The liver as an endocrine organ-linking NAFLD and insulin resistance . Endocr Rev . 2019 ; 40 : 1367 - 1393 . Google Scholar Crossref Search ADS PubMed WorldCat 100 Cantley J . The control of insulin secretion by adipokines: current evidence for adipocyte-beta cell endocrine signalling in metabolic homeostasis . Mamm Genome . 2014 ; 25 : 442 - 454 . Google Scholar Crossref Search ADS PubMed WorldCat 101 Rogal J , Binder C, Kromidas E, et al. WAT-on-a-chip integrating human mature white adipocytes for mechanistic research and pharmaceutical applications . Sci Rep . 2020 ; 10 : 1 - 12 . Google Scholar Crossref Search ADS PubMed WorldCat 102 Pedersen BK , Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ . Nat Rev Endocrinol . 2012 ; 8 : 457 - 465 . Google Scholar Crossref Search ADS PubMed WorldCat 103 Mizgier ML , Casas M, Contreras-Ferrat A, Llanos P, Galgani JE. Potential role of skeletal muscle glucose metabolism on the regulation of insulin secretion . Obes Rev . 2014 ; 15 : 587 - 597 . Google Scholar Crossref Search ADS PubMed WorldCat 104 Li X , Easley CJ. Microfluidic systems for studying dynamic function of adipocytes and adipose tissue . Anal Bioanal Chem . 2018 ; 410 : 791 - 800 . Google Scholar Crossref Search ADS PubMed WorldCat 105 Li X , Brooks JC, Hu J, Ford KI, Easley CJ. 3D-templated, fully automated microfluidic input/output multiplexer for endocrine tissue culture and secretion sampling . Lab Chip . 2017 ; 17 : 341 - 349 . Google Scholar Crossref Search ADS PubMed WorldCat 106 Lu S , Dugan CE, Kennedy RT. Microfluidic Chip with integrated electrophoretic immunoassay for investigating cell-cell interactions . Anal Chem . 2018 ; 90 : 5171 - 5178 . Google Scholar Crossref Search ADS PubMed WorldCat 107 Lim GE , Huang GJ, Flora N, LeRoith D, Rhodes CJ, Brubaker PL. Insulin regulates glucagon-like peptide-1 secretion from the enteroendocrine L cell . Endocrinology . 2009 ; 150 : 580 - 591 . Google Scholar Crossref Search ADS PubMed WorldCat 108 Liu Y , Kongsuphol P, Chiam SY, et al. Adipose-on-a-chip: a dynamic microphysiological in vitro model of the human adipose for immune-metabolic analysis in type II diabetes . Lab Chip . 2019 ; 19 : 241 - 253 . Google Scholar Crossref Search ADS PubMed WorldCat 109 Inge TH , Courcoulas AP, Jenkins TM, et al. Weight loss and health status 3 years after bariatric surgery in adolescents . N Engl J Med . 2015 ; 374 : 113 - 123 . Google Scholar Crossref Search ADS PubMed WorldCat 110 Rehfeld JF . The origin and understanding of the incretin concept . Front Endocrinol (Lausanne) . 2018 ; 9 : 1 - 7 . Google Scholar Crossref Search ADS PubMed WorldCat 111 Deacon CF . Peptide degradation and the role of DPP-4 inhibitors in the treatment of type 2 diabetes . Peptides . 2018 ; 100 : 150 - 157 . Google Scholar Crossref Search ADS PubMed WorldCat 112 Nguyen DTT , Van Noort D, Jeong IK, et al. Endocrine system on chip for a diabetes treatment model . Biofabrication . 2017 ; 9 : 015021 . Google Scholar Crossref Search ADS PubMed WorldCat 113 Guida C , Ramracheya R. PYY, a therapeutic option for type 2 diabetes? Clin Med Insights Endocrinol Diabetes . 2020 ; 13 : 117955141989298 . Google Scholar Crossref Search ADS WorldCat 114 Luo JZQ , Xiong F, Al-Homsi AS, et al. Human BM stem cells initiate angiogenesis in human islets in vitro . Bone Marrow Transplant . 2011 ; 46 : 1128 - 1137 . Google Scholar Crossref Search ADS PubMed WorldCat 115 Ellis CE , Vulesevic B, Suuronen E, Yeung T, Seeberger K, Korbutt GS. Bioengineering a highly vascularized matrix for the ectopic transplantation of islets . Islets . 2013 ; 5 : 216 - 225 . Google Scholar Crossref Search ADS PubMed WorldCat 116 Takahashi Y , Sekine K, Kin T, Takebe T, Taniguchi H. Self-condensation culture enables vascularization of tissue fragments for efficient therapeutic transplantation . Cell Rep . 2018 ; 23 : 1620 - 1629 . Google Scholar Crossref Search ADS PubMed WorldCat 117 Rambøl MH , Han E, Niklason LE. Microvessel network formation and interactions with pancreatic islets in three-dimensional Chip cultures . Tissue Eng Part A . 2020 ; 26 : 556 - 568 . Google Scholar Crossref Search ADS PubMed WorldCat 118 Becker MW , Simonovich JA, Phelps EA. Engineered microenvironments and microdevices for modeling the pathophysiology of type 1 diabetes . Biomaterials . 2019 ; 198 : 49 - 62 . Google Scholar Crossref Search ADS PubMed WorldCat 119 Wimmer RA , Leopoldi A, Aichinger M, et al. Human blood vessel organoids as a model of diabetic vasculopathy . Nature . 2019 ; 565 : 505 - 510 . Google Scholar Crossref Search ADS PubMed WorldCat 120 Edington CD , Chen WLK, Geishecker E, et al. Interconnected microphysiological Systems for Quantitative Biology and Pharmacology Studies . Sci Rep . 2018 ; 8 : 1 - 18 . Google Scholar Crossref Search ADS PubMed WorldCat 121 Miller PG , Shuler ML. Design and demonstration of a pumpless 14 compartment microphysiological system . Biotechnol Bioeng . 2016 ; 113 : 2213 - 2227 . Google Scholar Crossref Search ADS PubMed WorldCat 122 Arnold SE , Arvanitakis Z, Macauley-Rambach SL, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums . Nat Rev Neurol . 2018 ; 14 : 168 - 181 . Google Scholar Crossref Search ADS PubMed WorldCat 123 Shirakawa J , De Jesus DF, Kulkarni RN. Exploring inter-organ crosstalk to uncover mechanisms that regulate β-cell function and mass . Eur J Clin Nutr . 2017 ; 71 : 896 - 903 . Google Scholar Crossref Search ADS PubMed WorldCat 124 Hussain MA , Akalestou E, jin SW. Inter-organ communication and regulation of beta cell function . Diabetologia . 2016 ; 59 : 659 - 667 . Google Scholar Crossref Search ADS PubMed WorldCat 125 Eglen R , Reisine T. Primary cells and stem cells in drug discovery: emerging tools for high-throughput screening . Assay Drug Dev Technol . 2011 ; 9 : 108 - 124 . Google Scholar Crossref Search ADS PubMed WorldCat 126 Eglen RM , Randle DH. Drug discovery goes three-dimensional: goodbye to flat high-throughput screening? Assay Drug Dev Technol . 2015 ; 13 : 262 - 265 . Google Scholar Crossref Search ADS PubMed WorldCat 127 Shirakawa J , Kulkarni RN. Novel factors modulating human β-cell proliferation . Diabetes Obes Metab . 2016 ; 18 : 71 - 77 . Google Scholar Crossref Search ADS PubMed WorldCat 128 Shen W , Tremblay MS, Deshmukh VA, et al. Small-molecule inducer of β cell proliferation identified by high-throughput screening . J Am Chem Soc . 2013 ; 135 : 1669 - 1672 . Google Scholar Crossref Search ADS PubMed WorldCat 129 Wang W , Walker JR, Wang X, et al. Identification of small-molecule inducers of pancreatic β-cell expansion . Proc Natl Acad Sci USA . 2009 ; 106 : 1427 - 1432 . Google Scholar Crossref Search ADS PubMed WorldCat 130 Robitaille K , Rourke JL, McBane JE, et al. High-throughput functional genomics identifies regulators of primary human beta cell proliferation . J Biol Chem . 2016 ; 291 : 4614 - 4625 . Google Scholar Crossref Search ADS PubMed WorldCat 131 Walpita D , Hasaka T, Spoonamore J, et al. A human islet cell culture system for high-throughput screening . J Biomol Screen . 2012 ; 17 : 509 - 518 . Google Scholar Crossref Search ADS PubMed WorldCat 132 Mei Y , Hollister-Lock J, Bogatyrev SR, et al. A high throughput micro-array system of polymer surfaces for the manipulation of primary pancreatic islet cells . Biomaterials . 2010 ; 31 : 8989 - 8995 . Google Scholar Crossref Search ADS PubMed WorldCat 133 Desbordes SC , Studer L. Adapting human pluripotent stem cells to high-throughput and high-content screening . Nat Protoc . 2013 ; 8 : 111 - 130 . Google Scholar Crossref Search ADS PubMed WorldCat 134 Engle SJ , Vincent F. Small molecule screening in human induced pluripotent stem cell-derived terminal cell types . J Biol Chem . 2014 ; 289 : 4562 - 4570 . Google Scholar Crossref Search ADS PubMed WorldCat 135 Thakur G , Lee H, Jeon R, et al. Small molecule-induced pancreatic β-like cell development: mechanistic approaches and available strategies . Int J Mol Sci . 2020 ; 21 : 2388 . Google Scholar Crossref Search ADS WorldCat 136 Zhu Y , Liu Q, Zhou Z, et al. PDX1, Neurogenin-3, and MAFA: critical transcription regulators for beta cell development and regeneration . Stem Cell Res Ther . 2017 ; 8 : 1 - 7 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 137 Chen S , Borowiak M, Fox JL, et al. A small molecule that directs differentiation of human ESCs into the pancreatic lineage . Nat Chem Biol . 2009 ; 5 : 258 - 265 . Google Scholar Crossref Search ADS PubMed WorldCat 138 Kimura A , Toyoda T, Nishi Y, Nasu M, Ohta A, Osafune K. Small molecule AT7867 proliferates PDX1-expressing pancreatic progenitor cells derived from human pluripotent stem cells . Stem Cell Res . 2017 ; 24 : 61 - 68 . Google Scholar Crossref Search ADS PubMed WorldCat 139 Yuan Y , Hartland K, Boskovic Z, et al. A small-molecule inducer of pdx1 expression identified by high-throughput screening . Chem Biol . 2013 ; 20 : 1513 - 1522 . Google Scholar Crossref Search ADS PubMed WorldCat 140 Hill JA , Szabat M, Hoesli CA, et al. A multi-parameter, high-content, high-throughput screening platform to identify natural compounds that modulate insulin and Pdx1 expression . PLoS One . 2010 ; 5 : e12958 . Google Scholar Crossref Search ADS PubMed WorldCat 141 Szabat M , Modi H, Ramracheya R, et al. High-content screening identifies a role for Na+ channels in insulin production . R Soc Open Sci . 2015 ; 2 : 150306 . Google Scholar Crossref Search ADS PubMed WorldCat 142 Hohmeier HE , Zhang L, Taylor B, et al. Identification of a small molecule that stimulates human β-cell proliferation and insulin secretion, and protects against cytotoxic stress in rat insulinoma cells . PLoS One . 2020 ; 15 : 1 - 18 . Google Scholar Crossref Search ADS WorldCat 143 Stephens SB , Edwards RJ, Sadahiro M, et al. The Prohormone VGF regulates β cell function via insulin secretory granule biogenesis . Cell Rep . 2017 ; 20 : 2480 - 2489 . Google Scholar Crossref Search ADS PubMed WorldCat 144 Chung K , Wallace J, Kim SY, et al. Structural and molecular interrogation of intact biological systems . Nature . 2013 ; 497 : 332 - 337 . Google Scholar Crossref Search ADS PubMed WorldCat 145 Chen YY , Silva PN, Syed AM, Sindhwani S, Rocheleau JV, Chan WCW. Clarifying intact 3D tissues on a microfluidic chip for high-throughput structural analysis . Proc Natl Acad Sci USA . 2016 ; 113 : 14915 - 14920 . Google Scholar Crossref Search ADS PubMed WorldCat 146 Yamashita-Sugahara Y , Matsumoto M, Ohtaka M, et al. An inhibitor of fibroblast growth factor receptor-1 (FGFR1) promotes late-stage terminal differentiation from NGN3+ pancreatic endocrine progenitors . Sci Rep . 2016 ; 6 : 1 - 14 . Google Scholar Crossref Search ADS PubMed WorldCat 147 Zhang Y , Alexander M, Yang S, et al. High-throughput screening of encapsulated islets using wide-field lens-free on-Chip imaging . ACS Photonics . 2018 ; 5 : 2081 - 2086 . Google Scholar Crossref Search ADS WorldCat 148 Bitar CME , Markwick KE, Treľová D, et al. Development of a microchannel emulsification process for pancreatic beta cell encapsulation . Biotechnol Prog . 2019 ; 35 : 1 - 12 . Google Scholar Crossref Search ADS WorldCat 149 Delcassian D , Luzhansky I, Spanoudaki V, et al. Magnetic retrieval of encapsulated Beta cell transplants from diabetic mice using dual-function MRI visible and retrievable microcapsules . Adv Mater . 2020 ; 32 : 1904502 . Google Scholar Crossref Search ADS WorldCat 150 Köllmer M , Appel AA, Somo SI, Brey EM. Long-term function of alginate-encapsulated islets . Tissue Eng Part B Rev . 2016 ; 22 : 34 - 46 . Google Scholar Crossref Search ADS PubMed WorldCat 151 Liu H , Wang Y, Wang H, et al. A droplet microfluidic system to fabricate hybrid capsules enabling stem cell Organoid engineering . Adv Sci . 2020 ; 7 ( 11 ): 1903739 . Google Scholar Crossref Search ADS WorldCat 152 Amin J , Ramachandran K, Williams SJ, Lee A, Novikova L, Stehno-Bittel L. A simple, reliable method for high-throughput screening for diabetes drugs using 3D β-cell spheroids . J Pharmacol Toxicol Methods . 2016 ; 82 : 83 - 89 . Google Scholar Crossref Search ADS PubMed WorldCat 153 Walker SL , Ariga J, Mathias JR, et al. Automated reporter quantification in vivo: high-throughput screening method for reporter-based assays in zebrafish . PLoS One . 2012 ; 7 : e29916 . Google Scholar Crossref Search ADS PubMed WorldCat 154 Helker CSM , Mullapudi ST, Mueller LM, et al. A whole organism small molecule screen identifies novel regulators of pancreatic endocrine development . Development . 2019 ; 146 : dev172569 . Google Scholar PubMed OpenURL Placeholder Text WorldCat 155 Wang G , Rajpurohit SK, Delaspre F, et al. First quantitative high-throughput screen in zebrafish identifies novel pathways for increasing pancreatic β-cell mass . Elife . 2015 ; 4 : 1 - 26 . Google Scholar OpenURL Placeholder Text WorldCat 156 Tsuji N , Ninov N, Delawary M, et al. Whole organism high content screening identifies stimulators of pancreatic Beta-cell proliferation . PLoS One . 2014 ; 9 : e104112 . Google Scholar Crossref Search ADS PubMed WorldCat 157 Oberhauser L , Granziera S, Colom A, et al. Palmitate and oleate modify membrane fluidity and kinase activities of INS-1E β-cells alongside altered metabolism-secretion coupling . Biochim Biophys Acta—Mol Cell Res . 2020 ; 1867 : 118619 . Google Scholar Crossref Search ADS PubMed WorldCat 158 Duan H , Li Y, Arora D, Xu D, Lim HY, Wang W. Discovery of a benzamide derivative that protects pancreatic β-cells against endoplasmic reticulum stress . J Med Chem . 2017 ; 60 : 6191 - 6204 . Google Scholar Crossref Search ADS PubMed WorldCat 159 Tran K , Li Y, Duan H, Arora D, Lim HY, Wang W. Identification of small molecules that protect pancreatic β cells against endoplasmic reticulum stress-induced cell death . ACS Chem Biol . 2014 ; 9 : 2796 - 2806 . Google Scholar Crossref Search ADS PubMed WorldCat 160 Hrvatin S , O'Donnell CW, Deng F, et al. Differentiated human stem cells resemble fetal, not adult, β cells . Proc Natl Acad Sci . 2014 ; 111 : 3038 - 3043 . Google Scholar Crossref Search ADS PubMed WorldCat 161 Kalwat MA , Wichaidit C, Nava Garcia AY, et al. Insulin promoter-driven Gaussia luciferase-based insulin secretion biosensor assay for discovery of β-cell glucose-sensing pathways . ACS Sens . 2016 ; 1 : 1208 - 1212 . Google Scholar Crossref Search ADS PubMed WorldCat 162 Lee SH , Cunha D, Piermarocchi C, et al. High-throughput screening and bioinformatic analysis to ascertain compounds that prevent saturated fatty acid-induced β-cell apoptosis . Biochem Pharmacol . 2017 ; 138 : 140 - 149 . Google Scholar Crossref Search ADS PubMed WorldCat 163 Chen Z , Wang J, Sun W, et al. Synthetic beta cells for fusion-mediated dynamic insulin secretion . Nat Chem Biol . 2018 ; 14 : 86 - 93 . Google Scholar Crossref Search ADS PubMed WorldCat 164 Ribeiro D , Kvist AJ, Wittung-Stafshede P, et al. 3D-models of insulin-producing β-cells: from primary islet cells to stem cell-derived islets . Stem Cell Rev Rep . 2017 ; 14 : 177 - 188 . Google Scholar Crossref Search ADS WorldCat 165 Smink AM , de Vos P. Therapeutic strategies for modulating the extracellular matrix to improve pancreatic islet function and survival after transplantation . Curr Diab Rep . 2018 ; 18 : 39 . Google Scholar Crossref Search ADS PubMed WorldCat Author notes Funding information Allen Foundation; Eunice Kennedy Shriver National Institute of Child Health and Human Development, Grant/Award Number: P01 HD093363; National Institute of Diabetes and Digestive and Kidney Diseases, Grant/Award Number: UG3 DK119982; Shipley Foundation © AlphaMed Press 2021 This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model) TI - Engineering-inspired approaches to study β-cell function and diabetes JF - Stem Cells DO - 10.1002/stem.3340 DA - 2021-05-01 UR - https://www.deepdyve.com/lp/oxford-university-press/engineering-inspired-approaches-to-study-cell-function-and-diabetes-kQzaz5djbK SP - 522 EP - 535 VL - 39 IS - 5 DP - DeepDyve ER -