Introduction

In early 1869, with his mention of “small cells of almost perfect homogeneous content, and of perfect polygonal form … lying together in twos or in small groups,” [1] Paul Langerhans quietly announced to the medical community the discovery of what would later become known as the “islets of Langerhans”. This announcement would also set in motion the groundwork for successfully treating diabetes mellitus [2, 3]. At the time of the discovery, the function of these small clusters of cells was unknown, and Langerhans did not speculate about it. Twenty years would elapse between Langerhan’s initial discovery and the connection between the pancreas (and by default, islets) and diabetes was clearly established by Oskar Minkowski and Josef von Mering in 1889 [4]. By 1893, Gustave Édouard Laguesse hypothesized that the islets of Langerhans produced “internal secretions” that Jean de Meyer named “insulin” in 1909 [5]. Attempts to isolate this “internal secretion” began as early as 1907 but were not successful until the 1920s when insulin was first used to treat diabetes in a human patient [2, 5].

It was hypothesized that the exocrine tissue would impair the vitality and function of the endocrine pancreas [5, 6]. In 1902, Leonid W. Ssobolew suggested that the separation of the exocrine pancreas from the endocrine would increase islet viability and improve success in transplantation procedures [5, 6]. To this end, R. R. Bensley pioneered the staining of islets using neutral red and hand picking [5, 7]. But here the isolation process languished until 1964, when Claes Hellerström developed a technique using microscope microdissection [8]. In swift succession, beginning in 1965, isolation techniques evolved using collagenase [9], utilizing the anatomy of the pancreas in 1967 [10], and a density gradient in 1969 [11]. These three techniques were finally pulled together in 1985 by Gotoh et al [12] and truly opened the way for islet transplantation and in-depth study of islets.

Figure 1 shows this flow of advances in islet isolation. Since that time, there have been no major changes in the basic approach to islet isolation procedures. Our lab provided “A Practical Guide to Rodent Islet Isolation”, describing our insights into these procedures in 2009 [13]. Other groups have looked at a variety of digestive enzymes [14,15,16], variations on delivery of digestive enzyme [14, 17], and the separation of islets from exocrine tissue [18,19,20]. Some researchers have advocated the use of filtration to purify islets, skipping the density gradient completely [21, 22]. There have also been efforts to use stem cells [23,24,25] or dissociated islet cells and human amniotic epithelial cells [26], thus attempting to reduce or eliminate the need for islet isolations.

Fig. 1
figure 1

Timeline of advances in the understanding of islets of Langerhans

Islet isolation was pioneered in a very narrow range of species, specifically guinea pigs [7, 9,10,11, 27, 28] and mice [8, 12], with mice and rats still being the most often used species [16, 29,30,31]. Pigs [25], goats [32], cats [33], monkeys [34, 35], and dogs [36, 37] are also used to study various aspects of diabetes and potential treatments.

The primary goal of islet isolation, whether for in vitro studies or for transplantation, is to obtain purified islets that are both viable and responsive to stimulation in a manner consistent with their function in vivo. To this end, we highlight three key elements of a successful islet isolation procedure: 1) Digesting the tissues connecting the islets to the exocrine tissue, 2) separating islets from non-islet tissue, and 3) culturing islets in an environment that maintains viability and function (Fig. 2). This paper will review these key elements and provide detailed methods used in our laboratory to consistently procure viable and functional islets for research. Our protocols are by no means the only successful methods for the isolation of pancreatic islets; however, the additional details provided in these protocols for each step will hopefully assist researchers in their efforts to obtain a large number of healthy islets for both study and transplantation.

Fig. 2
figure 2

A graphical representation of the three key elements for a successful islet isolation. (1) The successful dissection of the pancreas followed by digestion. (2) Separation of the islets from acinar tissue using a separation gradient. It should be noted that islets are not visible to the naked eye and are thus not to scale in the illustration. (3) Culturing islets to ensure viability and functionality

Procedures of Islet Isolation

Basic Methodologies

When assessing and evaluating any islet isolation protocol consideration must be given to differences in the type and concentration of collagenase, the method of administration of the collagenase, temperature and duration of the digestion phase, the method used to purify islets from pancreatic acinar tissue, and the culture conditions following isolation. Each of these considerations are discussed below. While these items were discussed in our previous guide [13], each has been expanded, providing more up to date information for those who are new to the study of islets.

A survey of the literature for rodent islet isolation reveals a variety of methods for delivering the digestive enzymes to the pancreatic tissue surrounding the islets as well as different digestive enzymes. One method of delivery is to excise the pancreas from a euthanized animal and cut it into pieces, increasing the exposed surface area and providing improved conditions for the digestive enzyme to break down the tissue surrounding the islets [10, 17, 38]. The pieces are then enzymatically digested in the solution while also being mechanically digested by either shaking or stirring. A second method, described by Gotoh et al, has collagenase injected into the common bile duct of a euthanized animal. The inflated pancreas is then excised and digested at 37 °C without being cut into pieces or the addition of mechanical digestion [39].

While these two approaches form the foundation of many islet isolation techniques, there are considerable variations among published methods as well as alternative methods [8, 16, 20, 29, 40,41,42,43]. The advantages of using the common bile duct are twofold: 1) it allows the digestive enzyme to access the islets using the anatomical structures of the pancreas, and 2) stationary digestion reduces mechanical damage to the islets. Our laboratory uses a modification of the common bile duct protocol when isolating rodent islets using collagenase (see Appendix A for an annotated murine protocol and Appendix B for a bullet point protocol). Although cannulation of the murine bile duct requires technical skill, we are partial to this approach, as the collagenase interacts more closely with the connective tissue surrounding the islets when it is delivered through the intact pancreatic ductal system [44,45,46], resulting in a greater number of successfully isolated islets. There are several detailed online video accounts of a rodent islet isolation using a method of bile duct cannulation [16, 29, 30, 47,48,49].

Collagenase – Selection and Use

Collagenases are proteolytic enzymes that digest collagen, a major structural protein in animals [50, 51] and have been found in everything from bacteria to mammals [50,51,52,53]. Out of all the potential sources, it was found that all collagens or collagen-like proteins are susceptible to the enzyme obtained from Clostridium histolyticum (Cl. histolyticum) [51], the bacterium from which collagenase was first purified in the 1950s [54]. Subsequent studies found the raw collagenase obtained from this bacterium is a mixture of several different collagenases distinguished by molecular mass, termed alpha, beta, gamma, delta, epsilon, zeta, and eta [55, 56]. These have been at least partially characterized for stability and specificity [54, 55, 57,58,59,60,61]. It was also found that these collagenases could be separated into two distinct classes based on substrate specificity and amino acid analysis [58]. Class I collagenase encompasses the isoforms designated alpha, beta, gamma, and eta [56]. These four have high collagenase activity and moderate activity on the synthetic peptide 2-furanacryloyl-L-leucylglycyl-L-prolyl-L-alanine (FALGPA) [62]. The class II collagenases are delta, epsilon, and zeta [56] and have moderate collagenase activity and high FALGPA activity [62]. After these classifications were made, it was found that Cl. histolyticum has two distinct, yet separate genes that code for collagenase. The gene designated colG codes for those collagenases in class I while the colH gene codes for class II collagenases [63]. Since it is difficult to completely purify one type of collagenase from another, and the enzymes act synergistically, characterization is of vital importance.

Since collagenase is purified from bacterial culture, considerable variability exists between manufacturers as well as between production lots of collagenase from the same manufacturer. Therefore, the formulation, enzyme activity and purity of a given lot all strongly influence the outcome of any islet isolation. The composition of the collagenase and the concentration of enzymes in a specific lot must therefore be ideally suited to the task of islet isolation. A detailed description of the differences in isolation with purified collagenase type I and II as well as the combination that yields the most effective isolation of rat islets have been described by Wolters et al [64, 65]. Others formulated criteria for evaluating each lot of commercially available collagenase to ensure proper digestion of rat islets [66]. Those collagenase formulations with increased collagenase activity, low levels of trypsin activity, and a specific range of both neutral proteases and clostripain may yield the most viable islets [64, 66]. Our lab has found that optimal collagenase formulations used for rat islet isolation also provide acceptable criteria for rating collagenase used in our mouse islet isolation procedure.

Thus, there is a remarkable range of digestive enzyme formulations available for islet isolation, from crude collagenases to highly purified combinations used extensively for human islet transplantation. It has been suggested that the differences from lot to lot may be due to the lack of proper measurement of trypsin-like activity, even in the highly purified mixtures used in the isolation of human islets for transplantation [67]. Brandhorst et al suggested the trypsin-like activity in enzyme blends may work in concert with the other enzymes to increase the activity of the digestion, but there is some debate about the damage the trypsin-like activity has on the islets [68]. To ensure consistent activity and reproducibility for human islet isolation, the enzyme blends with both high purity and a precise notation of components are used, such as collagenase NB1 (Nordmark, Uetersen, Germany), Liberase (Roche, Basel, Switzerland) and Vitacyte HA (Vitacyte, Indianapolis, IN, USA) [67, 69,70,71]. These higher purity, and consequently high priced, enzymes are also used in human islet isolation to reduce the incidence of contamination by endotoxins [71, 72].

It has been shown that the presence of endotoxins correlates with an increase of the proinflammatory cytokines interleukin-1beta (IL-1beta), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-alpha) in both rat and human islets [69, 73, 74]. Both collagenase formulations and the different types of gradient compounds have been found to contain endotoxin in varying amounts [69]. Endotoxins are of particular concern in human islet transplant procedures especially since the infiltration of inflammatory cytokines in transplanted islets has been linked to endotoxin contamination in both enzyme blends and separation gradients [19, 69, 73, 74].

The Process of Collagenase Digestion

The process of collagenase digestion is of the utmost importance to the success of any given protocol. To ensure success, consideration must be given to the type of collagenase, its activity, the concentration of the collagenase solution, its route of administration, and the duration and temperature of the digestion. These factors can all vary widely among the various protocols. Clamping or tying off the ampulla of Vater where the hepatopancreatic duct connects to the duodenum (at the sphincter of Oddi) and perfusing the pancreas through the common bile duct allows collagenase to access the islets using the biological structures of the pancreas (Fig. 3). This method may change the duration of the digestion when compared to other methods. Differences in composition between lots of collagenase and other factors influence digestion as well. Thus, it is important to test each protocol for optimal viability and islet function, both of which remain paramount to the success and reproducibility of islet isolation.

Fig. 3
figure 3

Anatomy of the mouse upper intraperitoneal cavity showing the injection site and clamping of the common bile duct at the duodenum. NOTE: The top is the caudal portion of the mouse, bottom is the rostral portion, i.e., this is from the perspective of the mouse lying on its back tail away from the surgeon and nose toward the surgeon.

The duration of collagenase exposure is particularly important and should be optimized for each production lot. Figure 4 shows islets immediately post-separation when digestion time is not optimized to the unique collagenase lot. Digestion temperature, collagenase concentration, and route of administration were all the same; only digestion time was altered. In Fig. 4a, digestion time was not sufficient to separate the islets from the acinar tissue, resulting in clumping from which the islets will be difficult if not impossible to remove. These grossly encumbered islets will not be suitable for experiments. Figure 4b shows heavily over digested islets. Islets that have been over digested are fewer in number and typically either very large or very small. There also tends to be much finer debris in the plate as the islets will shed dead and dying cells for some time. In our experience, over digested islets are not as responsive to stimulation and do not survive overnight culture well.

Fig. 4
figure 4

Examples of both under digested and over digested islets immediately post isolation and purification. a Under digested islets will typically contain large amounts of acinar tissue appearing dark brown or black. Islets are often aggregated near the masses of acinar tissue as shown. Separating islets from these under digested clumps is often difficult. b Over digested islet preparations result in large amounts of pancreatic debris consisting of tiny fragments of acinar tissue and digested islets

Our lab, following published guidelines for collagenase enzyme formulations [66], uses Collagenase P (Roche, Basal, Switzerland, #11249002001) enzyme at 1.4 mg/mL in a modified Hank’s Balanced Salt Solution (HBSS) (Invitrogen, Carlsbad, CA, USA, #14065–056), injected into the pancreas via the common bile duct. The pancreas is then excised whole and placed in modified HBSS for stationary digestion at 37 °C for seven to 10 min as described in detail in Appendix A. Ideally, three separate digestions using two mice per time point should be completed to evaluate a newly purchased lot of collagenase. Our lab uses timepoints of 7, 8, and 9 min. After overnight incubation, islets from each digestion should be visually inspected and tested for viability and functionality using the assays reviewed below. The group with the highest yield (assuming proper cannulation) and best outcome in the assays indicates the proper digestion time.

Separation of Islets and Pancreatic Acinar Tissue Using a Density Gradient

Purifying islets from acinar tissue, regardless of the method used, is important due to the nature of pancreatic tissue. The cells of the exocrine pancreas secrete various digestive enzymes that negatively impact islet viability [75]. Our lab has used both Ficoll 400 (MilliporeSigma, Burlington, MA) and Histopaque (MilliporeSigma, Burlington, MA) at different densities. In isolations using Ficoll 400 layered in a discontinuous gradient of 1.109, 1.096, 1.070, and 0.570 g/ml, islets were recovered from the interfaces of both the 1.070/1.096 and 1.096/1.109 g/ml layers. We found, however, that the preparations were often contaminated with acinar tissue. Using aseptically filled and premixed Histopaque, a mixture of sodium diatrizoate and Ficoll 400, we found that our purity results generally improved. Combining Histopaque 1.119 g/mL with the 1.077 g/mL preparation to produce a 1.100 g/mL gradient appears to enhance islet purity. It should be noted that our studies comparing Histopaque and Ficoll were not rigorous, and both gradients are widely used and accepted. McCall et al compared Histopaque, Ficoll, Dextran and Iodixanol assessing recovery, viability, purity, and in vitro functionality [76].

Depending on how much residual acinar tissue remains following the initial purification of the islets, a second purification is often needed to further increase the purity prior to culture. Our protocol includes using a basic light microscope with a 4x objective to identify islets, then handpicking those islets from one suspension culture dish into a second culture dish. Ideally, this is done 2 h post isolation. The islets then recover overnight in a 37 °C incubator with 5% CO2. The following morning, the islets are again handpicked into a new dish, thus eliminating any acinar tissue that may have either been transferred with the initial cleaning or was attached to the islets after isolation. This also removes healthy islets from any buildup of toxins from dying acinar tissue and from any necrosing islets that did not survive. It should be stated that once the islets are transferred to media, minimizing time outside the temperature-controlled and sterile conditions of the incubator will limit contamination and pH changes while handpicking islets for experiments.

Islet Yields

Overall yield from a rodent pancreas is highly variable among strains. A comparison of seven different mouse strains was made by Bock et al and the number of islets per pancreas was found to range from ~ 1000 in 129S6 mice to ~ 2500 in B6 mice [77]. Using a mouse model of diabetes, they identified a similar number of islets per pancreas (~ 3200) for both the ob/ob and the ob/+ controls [78]. More recently, Mitok et al reported the average islet yield for eight different strains of mice, with > 400 islets/mouse from five of the strains (B6, A/J, WSB, CAST, and PWK), < 300 islets/mouse for the 129 and NOD strains, and notable sex differences in islet yield for the NZO strain [31]. De Haan et al investigated yields from different rat strains as well as factors that potentially influenced yields [66]. Inuwa et al demonstrated that the total number of islets in young male Wistar rats increased with age, ranging from ~ 6000 to ~ 20,000 islets per pancreas [79], while others have estimated the number per rat pancreas as low as ~ 3000–5000 islets [80, 81]. Thus, the expected islet yield from an isolation procedure depends a great deal on the age, sex, and strain of the rodent.

The expertise of the technician, as well as the method of isolation chosen, will also influence the total islet yield, making a definitive expected yield difficult to quantify. In our lab an experienced technician using 8–12-week-old CD-1 mice (Envigo, Indianapolis, IN) will usually obtain between 300 and 450 islets on average, while a similarly aged C57BL/6 J (Jackson Laboratory, Bar Harbor, ME) will yield only ~ 250–350 islets with an average yield of 300 islets per mouse. De Groot et al report rat yields of approximately 600–800 islets per animal [82]. Since the endocrine pancreas accounts for between 1 and 4% of the total volume of the pancreas [83], this suggests islet yields range from 10 to 30% from a typical rodent pancreas. For comparison, it is thought the human pancreas contains over one million islets, yields of approximately 250,000–300,000 islets are estimated by islet equivalents (IEQ) [84], although there are notable issues with IEQ quantification and no clear consensus on islet yields from human donors [85].

Conditions for Islet Culture

While our previous guide [13] contained information on conditions for islet culture, this guide has expanded that information as well as provided visual representations for islets at three different time points post-isolation. After an islet isolation, it is vital to ensure proper culture conditions so islets may recover from the process of collagenase digestion. An examination of media with different glucose concentrations showed that rat and mouse islets cultured with 11 mM glucose have lower apoptosis rates, increased viability, and increased immunoreactive insulin compared to islets cultured in either higher or lower glucose concentrations [86, 87]. Culture media with glucose concentrations substantially below 11 mM can reduce the insulin content in the islet as well as downregulate key genes related to glucose metabolism, whereas prolonged exposure to high glucose may lead to toxicity or other functional impacts [87,88,89]. Of all the culture media tested, RPMI 1640 supplemented with serum best maintained or augmented glucose-stimulated insulin secretion in murine islets [86]. Other groups have reported success in maintaining viability and function with rat islets in CMRL 1066 [66] and Iscove’s MEM [90].

Our lab uses RPMI 1640 (Invitrogen, Carlsbad, CA, USA #11875–093) culture media supplemented with 10% (v/v) fetal bovine serum (R&D Systems, Minneapolis, MN, USA #S11150) to promote viability and 100 U/mL penicillin and 100 mg/mL streptomycin (Invitrogen, Carlsbad, CA, USA #15140–122) to reduce contamination both for culturing of islets and while purifying islets from acinar tissue after density gradient separation. Islets are plated in 100 mm × 20 mm suspension culture dishes (Corning Inc., Corning, NY, #430591) as recommended [86], with 11 mL RPMI 1640 complete media in each dish.

An optimal density of four islets per square centimeter has been suggested to prevent competition for nutrients [66]. Using the standard 100 mm × 20 mm culture dish with ~55cm2, this allows for ~ 220 islets per culture dish. Our lab prepares three culture plates per mouse, one for the islets immediately post separation, one plate for the initial hand cleaning of the islets two to 4 h post separation, and the last plate for the final cleaning after overnight (16–20 h) recovery. The initial 2–4-h recovery period allows for the islets to rest from the insult of collagenase digestion and to begin recovery. Cleaning the islets from the acinar tissue and any dead or dying islets increases the chance of islet survival. The second cleaning the following morning should result in a pure islet culture with little or no acinar tissue. Each of these time points are shown in Fig. 5. Islets immediately post separation (Fig. 5a) have some acinar tissue still evident on the islets. The same islets are shown in Fig. 5b after a two-hour recovery period and initial hand cleaning. Figure 5c shows the same islets after overnight recovery and hand picking a final time. Note there is still some acinar tissue present in the final image.

Fig. 5
figure 5

Islets after optimal digestion and recovery time. a An example of islets immediately post-isolation and purification. b-c The same preparation of islets after a two-hour recovery period but before cleaning (b) and after overnight recovery and before a final cleaning (c). Note that 88 islets were counted in (b) and 80 islets in (c) even though the tissue density appears quite different between images. Pancreatic debris accounts for the additional tissue observed in (b) prior to final cleaning

To maintain optimal islet health and function, storage in a sterile incubator at 37 °C with 5% CO2 and a humidified atmosphere is necessary [86]. Rodent islets can maintain glucose sensitivity for at least 1 week in culture with frequent media changes [86] and perhaps even longer based on data from human islets [86, 91]. It should be noted, however, that changes in rodent islet function can occur in as little as one to 4 days in culture [92].

Dispersion of Islets into Islet Cells

Although the intact islet provides an excellent model system for studying the function of this pancreatic micro-organ, some experimental approaches require dispersion of islets into individual islet cell cultures. Examining subtypes of islet cells requires islet dispersion as a requisite step. Thus, islet dispersion allows for the separation and examination of islet subpopulations such as alpha, beta, delta, and other islet cell types. Further, dispersion provides a convenient model of the isolated beta cell by preventing gap junction coupling [93, 94], blocking calcium waves between cells [95], and reducing paracrine signaling [96, 97]. This allows for comparisons between intact islets and dispersed beta cells [98]. A detailed protocol for dispersing, culturing, and identifying islet cells in culture is described in in Scarl & Koch et al. [99].

Assessing Islet Health and Function

Once islets have been isolated and placed in culture, their health and function need to be assessed. In this section, we have updated and expanded the original description of assessment techniques presented in Carter et al [13].

Morphology

Rudimentary information regarding the health of an islet may be provided by visual inspection. When viewed under a light microscope with 4x magnification, healthy rodent islets appear spherical and golden-brown in color, with an approximate diameter of 50–250 μm. These features, particularly the color when compared to the exocrine tissue, allow for rapid identification of islets. Healthy isolated islets following overnight recovery have few, if any, individual cells protruding from their relatively smooth surface, such as the islet in Fig. 6a. Cells protruding from the surface of an islet (Fig. 6b) can be a sign of decreased or decreasing health, as is acinar tissue clinging to the exterior of an islet (Fig. 6b). Larger islets are also prone to developing hypoxic cells in their core, visibly distinguished as a darker region when compared to the surrounding area (Fig. 6c). It has been suggested that hypoxia can be reduced by culturing islets at a lower incubation temperature initially [100] or by reducing the volume of media in the culture dish to increase oxygenation [101].

Fig. 6
figure 6

Islets found in a typical preparation. a Example of a healthy islet with amber color. b Example of an islet with acinar tissue, which typically appears as dark brown or black tissue on the exterior of the islet. c Example of an islet with a necrotic core due to hypoxia

Viability

To provide more quantitative measures of islet health than visual inspection alone, several techniques can be used to assess islet viability. We define viability as living versus dead or dying cells as assessed using cell exclusion or DNA-binding dyes. A common approach that is widely used in the human islet transplant field is to estimate the ratio of healthy living cells to dead cells within each islet by fluorescence microscopy. Fluorescein diacetate (FDA) incorporates into healthy cells by facilitated diffusion and fluoresces blue, and propidium iodide (PI) is a membrane impermeant red fluorescent dye that is excluded from viable cells and enters only dead or dying cells [102]. Using these fluorescent dyes in combination, the health of islets can be assessed by the FDA/PI ratio. Unmanipulated islets isolated from healthy control animals generally have 90–95% viability, meaning blue FDA staining in 90–95% of the component cells of an islet and red PI staining in only 5–10% of the cells in an islet. Additional techniques include AnnexinV (AnnV), SYTO-13/ethidium bromide, calcein AM/ethidium homodimer, fluorescein diacetate, and ethidium bromide [102,103,104].

Rodent islets in a good preparation are uniformly viable at > 95%, so we primarily focus on quantifying cell death only. We use a variation of the above approach by quantifying the mean intensity of AnnV fluorescence to measure apoptosis and PI fluorescence to measure generalized cell death within an islet (see Appendix C). A region of interest is drawn around the perimeter of each islet using imaging software (CellSens, Olympus, USA) to quantify the fluorescent intensity of cell death signal per unit of cross-sectional area for each islet. Figure 7 provides an example of typical staining patterns for cell death following overnight culture in normal conditions (6A, top), ER-stress inducer thapsigargin (6A, middle), and proinflammatory cytokines (6A, bottom). As shown in Fig. 7b, almost no AnnV staining was observed in controls (top), whereas substantial AnnV staining was observed in the other conditions. In Fig. 7c, virtually no fluorescence is detected in control islets (top), and only one region on one islet from the thapsigargin-treated group showed any PI staining (middle). In contrast, PI staining was extensive among cytokine-treated islets (Fig. 7c, bottom). These observations are consistent with established apoptosis-specific pathways of thapsigargin-induced cell death [105] versus apoptotic and necrotic pathways activated by cytokines [106, 107]. We have published this approach previously to measure islet cell death in response to proinflammatory cytokines [108, 109] and high concentrations of metformin [110]. It should be noted that isolated islets, having lost vascular connection, can become severely hypoxic. This results in necrosis, especially in the core of the islet. Identification and removal of these islets from experiments can help in acquiring more accurate experimental data (see Supplemental Fig. S1 for additional details).

Fig. 7
figure 7

An example of thapsigargin-induced and cytokine-induced cell death as measured by annexin V and propidium iodide fluorescence. a A brightfield image of healthy mouse islets (top), islets treated overnight with 100 nM thapsigargin to induce ER stress (middle), and islets treated overnight with 5 ng/mL IL-1beta and 10 ng/mL TNF-alpha (cytokines) to induce cell death (bottom). b Annexin V is depicted with green fluorescence and propidium iodide is depicted with red fluorescence (c)

Glucose-Stimulated Insulin Secretion (GSIS)

Insulin is crucial in regulating blood glucose, and reduced insulin secretion is a key feature of both type 1 and type 2 diabetes [111]. A fundamental property of islets is their capacity to regulate insulin release in direct response to changes in extracellular glucose concentrations. This ability defines islet function since insulin is produced and released only from islet beta-cells. There are other peptides produced by islet cells, such as glucagon, somatostatin, ghrelin, pancreatic polypeptide, as well as numerous “nonclassical” peptides [112]. These peptides are secreted in smaller amounts than insulin and are potentially more difficult to detect. Glucose-stimulated insulin secretion (GSIS) is thus a well-accepted measure of islet function and is considered the gold standard.

GSIS may be measured either by static conditions or by perfusing islets to measure the kinetics of insulin release in response to glucose. Each technique has its advantages and disadvantages which are reviewed elsewhere [113]. Typically, to measure functional GSIS, islets are cultured in a ‘low’ glucose concentration near 3 mM. This is to measure the amount of insulin secreted into the supernatant under ‘basal’ or ‘unstimulated’ conditions. Stimulated insulin release is then measured by exposing the islets to a higher glucose concentration such as 11 mM (half-maximal) or > 20 mM (maximal). In response to glucose stimulation, the time course of the islet response is biphasic, with a rapid spike in insulin (first phase) followed by a decline to a prolonged plateau (second phase) that remains for the duration of the stimulus. These are used to obtain a stimulation index (SI), the ratio of stimulated-to-basal insulin secretion. Healthy islets have an SI of 2–20 depending on several factors including strain, age, body weight, and glucose concentrations selected [82, 90].

One of the drawbacks to GSIS is the large number of islets required to conduct a thorough assessment. In recent years, the development of micro-plate assays using one islet per well have been used in an attempt to reduce the number of islets required for assessment, especially with regard to human islet transplantation [114]. Another point of contention is how best to control for variations in insulin secretion amongst islets to present the data obtained from a GSIS assay. In Slepchenko et al, we compared several normalization processes including total protein, insulin content, and islet area [115]. We concluded that normalization is not helpful in most cases as long as islets are within a size range of approximately +/− 50% of the mean because insulin secretion appears to be generally independent of islet size [115]. Among the normalization methods available, measuring the cross-sectional islet area was optimal for normalizing insulin secretion [115].

Intracellular Calcium Imaging

Another important method to assess islet function is measuring changes in intracellular calcium ([Ca2+]i) in response to glucose or other stimuli [116]. We have used the ratiometric probe Fura-2 AM with an epifluorescent microscope [98], but there are many other calcium probes with different excitation and emission spectra that are capable of detecting changes in [Ca2+]i including fluo-3, fluo-4, and fura red. Improved versions of some of these fluorophores have reduced leakage and photobleaching [117, 118]. Transfectable probes that target specific calcium-containing organelles such as the endoplasmic reticulum and mitochondria are also available [119, 120]. These more sophisticated probes require Forrester resonance energy transfer (FRET).

We and others have found that Fura-2 AM can provide a basic picture of islet function and dysfunction [121,122,123]. As shown in Fig. 8a, changes in [Ca2+]i can be assessed across multiple glucose steps to provide a detailed view of dose-dependent glucose stimulation. The 0 and 4 mM glucose concentrations are subthreshold, so there is little change in [Ca2+]i activity as measured by the ratio of fluorescence intensity from 340/380 nm excitation light. The step to 8 mM glucose produces a classic biphasic response: an initial first phase peak followed by a second phase plateau (Fig. 8a). The 12 mM step provides the maximum first phase response in this example set of islets. The [Ca2+]i levels continue to rise with each glucose step to maximum stimulation in 20 mM glucose. This ‘one-shot’ experiment provides enough data to produce a glucose dose-response curve for both 1st phase (Fig. 8b) and 2nd phase (Fig. 8c) [Ca2+]i. The procedures we used for [Ca2+]i imaging can be found in Scarl & Koch et al. [99].

Fig. 8
figure 8

Calcium response to glucose stimulation. a Calcium traces recorded at 10-s intervals during a series of increasing glucose steps (0, 4, 8, 12, 16, 20 mM) in an 90-min recording (N=20 islets). For each glucose step both 1st phase (peak) and 2nd phase (plateau, measured in the last 5-min) were measured. b-c Mean values for each phase at each glucose step are used to assemble glucose dose-response curves for 1st phase (b) and 2nd phase (c) calcium responses as an indicator of islet function

The Use of Fluorescent Labels in [Ca2+]i Experiments

To extend the value of [Ca2+]i imaging as an evaluation tool of islet function, we utilize Cell Tracker Red (CTR) as a means to record and compare test groups side-by-side with control groups of islets. The basic approach was first described by Corbin et al. [124] and is shown in cartoon form in Fig. 9. Briefly, Fura-2 is diluted in one well of a 12-well plate (A), half of the solution is moved to an adjacent well, and CTR is added to the second well (B). Untreated or control islets are added to the first well while the ‘test islets’ (e.g. islets from a mutant mouse, islets treated with drugs to stimulate or inhibit insulin secretion, islets put under stress, etc.) are added to the second well and incubated for 30 min (C). At the end of the 30-min incubation (dye loading period) the islets are briefly combined in the Fura-2 only well (D) and then transferred to the recording chamber of the microscope. It should be noted that this will introduce a very small amount of CTR into the Fura-2 only well. This concentration is both extremely small and transient for the Fura-2 only islets since the fluid flow of the chamber will wash excess CTR and Fura-2 away. Islets are then located and imaged using brightfield illumination (E), then the red fluorescence is imaged from the CTR-labeled test cells only (F), and the [Ca2+]i signal from all islets is imaged (G). Data sets can be separated based on the red fluorescence image, with islets that are not visible belonging to the well 1 control and islets with red fluorescence belonging to the well 2 test condition.

Fig. 9
figure 9

Fluorescence imaging with Fura-2 and CTR. Fura-2 is diluted in one well (a), then a 1 ml aliquot is moved to an adjacent well and CTR is added (b). Islets are added to each well and incubated for 30 min (c) before being combined in the Fura-2 only well and transferred to the recording chamber (d). Islets are imaged using three different wavelengths: bright field (e), CTR only (f), and Fura-2 (g). Once the calcium imaging experiment is complete, the islets from the second well can be identified as the red islets in the CTR image

This CTR-labeling approach is displayed in Fig. 10. Here we show brightfield (A), red fluorescence (B), and Fura-2 signal (C) for sets of islets labelled with or without CTR. In Fig. 10d, we show [Ca2+]i traces from a set of islets that were isolated from a diabetic db/db mouse and loaded with CTR and Fura-2. In Fig. 10e, we show [Ca2+]i traces from the heterozygous controls that were loaded with Fura-2 only. As shown in Fig. 10f, the average responses were quite different, but the x- and y-axes accurately reflect the signal from both sets of islets. Recording control and test groups simultaneously controls for temperature, perifusion rate, and other variables, which allows us to identify subtle changes in islet function that are important when making statistical comparisons. Previous studies using this approach have identified signs of endoplasmic reticulum stress [109] and dissociations between insulin secretion and [Ca2+]i [125] by examining the latency, amplitude, and slope of islet [Ca2+]i responses to glucose stimulation. Although [Ca2+]i can be a convenient and insightful indicator of islet health and function, it is important to also measure insulin secretion due to many facets of the insulin secretory pathway that are separate from changes in [Ca2+]i [125,126,127,128].

Fig. 10
figure 10

Example of Cell Tracker Red (CTR) being used to identify two different treatment groups of islets. a-c Images of islets: group 1 contains control islets loaded with Fura-2 for 30 min, and group 2 contains “test” islets loaded with Fura-2 and 200 nM CTR for 30 min. Both groups of islets are combined in the recording chamber for live-cell fluorescence microscopy. a A brightfield image shows all islets (group 1 and group 2). b An image taken with filters to detect only red fluorescence light (535 nm Ex; 610 nm Em). Test islets (group 2) produce detectable red fluorescence due to the CTR label. Yellow circles surround the location of “invisible islets” from group 1 that were not labeled with CTR. c Fluorescence from the Fura-2 signal (380 nm Ex) is detectable in all islets, so that calcium recordings of group 1 and group 2 can be made simultaneously under identical experimental conditions. d-f An example of a calcium recording using CTR to compare diabetic and healthy islets. d Calcium traces from diabetic islets isolated from db/db mice loaded with 1uM Fura-2 + 200 nM CTR for 30 min. e Calcium traces from healthy islets isolated from heterozygous control mice loaded with 1uM Fura-2 only. f Mean calcium traces for db/db (N=5 islets) and heterozygous controls (N=5) can be used to assess differences in amplitude, slope, and latency by directly comparing two treatment groups under identical environmental conditions

Conclusions

As stated previously, islet isolation is an intricate process, but the ability to consistently procure viable and functional islets is crucial to effectively study the physiology and pathophysiology of islets. In this review, we have addressed the key factors to consider in both the isolation and assessment processes to obtain healthy islets. We provide a method of isolation developed by integrating the reports of many others in the research field with careful experimentation to optimize the islet isolation process for our laboratory. While this protocol provides a start for islet isolation, any procedure must be optimized to the capabilities of the laboratory and the specific goals of the study.