74 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 1 Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia 2 Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia 3 Institute for physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia Correspondence/ Korespondenca: Marko Milojević, e: marko. milojevic1@um.si Key words: in vitro models; pancreas; bioscaffolds; 3D printing; tissue slice method Ključne besede: modeli in vitro; trebušna slinavka; bionosilci; 3D tisk; metoda tkivne rezine Received: 18. 10. 2019 Accepted: 12. 5. 2020 eng slo element en article-lang 10.6016/ZdravVestn.3001 doi 18.10.2019 date-received 12.5.2020 date-accepted Metabolic and hormonal disorders Metabolne in hormonske motnje discipline Review article Pregledni znanstveni članek article-type In vitro models of the endocrine pancreas Modeli in vitro endokrine trebušne slinavke article-title In vitro models of the endocrine pancreas Modeli in vitro endokrine trebušne slinavke alt-title in vitro models, pancreas, bioscaffolds, 3D printing, tissue slice method modeli in vitro, trebušna slinavka, bionosilci, 3D tisk, metoda tkivne rezine kwd-group The authors declare that there are no conflicts of interest present. Avtorji so izjavili, da ne obstajajo nobeni konkurenčni interesi. conflict year volume first month last month first page last page 2021 90 1 2 74 90 name surname aff email Marko Milojević 1 marko.milojevic1@um.si name surname aff Andraž Stožer 3 Uroš Maver 1,2 eng slo aff-id Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia Inštitut za biomedicinske vede, Medicinska fakulteta, Univerza v Mariboru, Maribor, Slovenija 1 Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia Katedra za farmakologijo, Medicinska fakulteta, Univerza v Mariboru, Maribor, Slovenija 2 Institute for physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia Inštitut za fiziologijo, Medicinska fakulteta, Univerza v Mariboru, Maribor, Slovenija 3 In vitro models of the endocrine pancreas Modeli in vitro endokrine trebušne slinavke Marko Milojević,1,2 Andraž Stožer,3 Uroš Maver1,2 Abstract Ambitions to develop artificial tissue substitutes, combined with the need to study underlying mechanisms of disease under controlled conditions, shortcomings of animal models, as well as ethical constraints, were the main driving forces behind the development of advanced in vi- tro models. These are defined as alternative experimental systems made by leveraging recent advances in tissue engineering and additive manufacturing that mimic tissue or organ level physiology in vitro. Simple in vitro models are already being used in many applications, how- ever, due to their many drawbacks, they incompletely mimic dynamic responses of native tis- sues. In order to construct functionally more relevant in vitro models, cells need to be grown in three-dimensional (3D) environments or bioscaffolds. Generally, bioscaffolds must recapitulate the microarchitecture, hierarchical structure, physical properties, and composition of native tissues. Nonetheless, progress towards building more complex models is hindered primarily by the diffusion of gases and nutrients into the constructs´ interior. Currently, 3D printing pres- ents the most promising solution for the production of advanced bioscaffolds, which resolve the above-mentioned limitations. In addition to the technique´s ability to simultaneously use multiple biocompatible materials, 3D printing enables material deposition with micrometer spa- tial resolution under cell-friendly conditions. The development of a functional in vitro pancreas model is governed by the desire to study diabetes aetiology and is one of the main goals of the in vitro modelling domain, which to date remains unfulfilled. Despite having some drawbacks, the tissue slice method presents the gold standard for basic and translational studies of the pancre- as, while the currently most advanced 3D fabricated in vitro pancreas models mimic only basic functions of the organ. The purpose of this review is to provide an overview of in vitro models with a focus on in vitro models of the endocrine pancreas. We will highlight different model types and fundamental elements which need to be considered when constructing a model. Emphasis will be placed on more complex 3D fabricated in vitro models, tissue slices, bioscaffold material properties, and the use of 3D printing for the fabrication of advanced bioscaffolds. We believe that the simultaneous development of advanced materials, micro-manufacturing technologies, and advanced cell culture methods presents a very promising approach towards the construc- tion of a functional in vitro pancreas model. Izvleček Želja po razvoju tkivnih nadomestkov, potrebe po preučevanju mehanizmov bolezni v kontroli- ranih pogojih, pomanjkljivosti živalskih modelov in etične omejitve so bile povod za razvoj pod- ročja naprednih modelov in vitro. Te definiramo kot alternativne eksperimentalne sisteme, ki z uporabo modernih tehnik tkivnega inženirstva in aditivne proizvodnje posnemajo strukturo in funkcionalnost tkiv ali organov in vitro. Enostavni modeli in vitro se že uporabljajo v številne namene, vendar zaradi mnogih pomanjkljivosti ne posnemajo dovolj dinamičnih odzivov izvor- nih tkiv. Za izgradnjo funkcionalno kompleksnejših modelov in vitro je celice potrebno gojiti v tridimenzionalnem (3D) okolju ali bioloških nosilcih. Splošno morajo biološki nosilci posnemati mikroarhitekturo, hierarhično strukturo, fizikalne lastnosti in sestavo izvornega tkiva. Toda na- predek k izgradnji kompleksnejših modelov omejuje predvsem difuzija plinov in hranil v notra- Slovenian Medical Journal 75 REVIEW ARTICLE In vitro models of the endocrine pancreas 1 Introduction The main purpose of the majority of biomedical research is to decipher the origin and molecular mechanisms of hu- man diseases with the goal of developing new or better preventive, diagnostic, and therapeutic approaches. For obvious rea- sons, basic research alone cannot be per- formed on humans, and animal models differ too much anatomically, physiolog- ically, and genetically from humans, so they often do not mimic critical aspects of human healthy or pathologically altered tissues and organs, especially not with the desired molecular resolution (1). The development of alternative models that mimic human anatomy and physiology in vitro is therefore urgently needed. In par- allel, the ultimate goal of the field of tis- sue engineering (TE) is the production of functional tissues and organs in vitro that could be used as substitutes to repair or replace the damaged and diseased human body parts (2-4). Individual TE research njost konstrukta. Tehnika 3D tiska je trenutno najobetavnejša rešitev za proizvodnjo naprednih bioloških nosilcev, ki odpravljajo te pomanjkljivosti. 3D tisk poleg zmožnosti sočasne uporabe več biološko kompatibilnih materialov omogoča nalaganje materiala z mikrometrsko prostorsko ločljivostjo pri pogojih, primernih celicam. Vse od zasnove področja modelov in vitro je zaradi že- lje po preučevanju nastanka sladkorne bolezni razvoj funkcionalnega modela trebušne slinavke eden od osrednjih, a še nedoseženih ciljev. Zlati standard za bazične in translacijske raziskave trebušne slinavke pa je kljub nekaterim pomanjkljivostim metoda tkivne rezine, medtem ko tre- nutni najnaprednejši 3D zgrajeni modeli trebušne slinavke in vitro posnemajo le osnovne funkci- je organa. Namen tega članka je predstavitev modelov in vitro s poudarkom na modelih trebušne slinavke. Predstavil bo tipe modelov in ključne elemente, ki jih je treba pri izgradnji upoštevati. Poudarek bo na kompleksnejših 3D zgrajenih modelih in vitro in tkivnih rezinah, materialnih la- stnostih bioloških nosilcev ter tehniki 3D tiska za izgradnjo naprednih bioloških nosilcev. Meni- mo, da je sočasni razvoj znanosti o materialih, mikroproizvodni tehnologiji in celičnih kulturah izjemno obetavna pot k izgradnji funkcionalnega modela trebušne slinavke in vitro. Cite as/Citirajte kot: Milojević M, Stožer A, Maver U. In vitro models of the endocrine pancreas. Zdrav Vestn. 2021;90(1–2):74–90. DOI: https://doi.org/10.6016/ZdravVestn.3001 Copyright (c) 2021 Slovenian Medical Journal. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License. studies focus mainly on the development of individual milestones of this process in order to achieve the ultimate goal (e.g. development of advanced materials, pro- duction methods and cellular resources, etc.) (5,6) and on the construction of sim- ple in vitro models that mimic the basic functions of tissues and organs in vivo. In order to achieve the principles of 3Rs – replacement, reduction and refinement in the face of ethical limitations and short- comings of animal models and the goals of tissue engineering to study the mecha- nisms of disease in controlled conditions, they led to the development of complex in vitro models. The field of advanced in vi- tro models lies at the intersection of TE, regenerative medicine, pathophysiology, advanced materials science, and additive manufacturing and focuses on mimicking the three-dimensional (3D) structure and functionality of tissues or organs in vitro. 76 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 2 In vitro models 2.1 Simple in vitro models Simple two-dimensional (2D) in vi- tro models are already being used for a number of purposes (Figure 1), such as the development and testing of new ther- apeutical substances and for pharma- cological and toxicological studies. For this purpose, transformed or immortal commercially available cell lines are most often used, but during prolonged cultiva- tion (increase in the number of cell divi- sions), they usually begin to differ genet- ically, epigenetically and physiologically from primary cells. For the construction of in vitro models, commercial cell lines are, therefore, the worst choice (7,8). In contrast, isolated primary cell cultures are the best candidates for use in in vitro models, as they best represent functional units of the native tissue. The construction of in vitro models based on primary cell cultures is limited mainly due to the diffi- cult availability and isolation of cells that, in addition, have difficulty proliferating in standard 2D cell cultures and have a short lifespan (i.e., Hayflick limit) (9). Recently, these problems have been overcome by us- ing induced pluripotent stem cells (iPSCs), which can be obtained from somatic cells by a dedifferentiation process (10). iPSCs have the ability to self-regenerate and dif- ferentiate into many mature cell types, making them extremely interesting for use in in vitro models where we study the on- set and the course of disease development. The reason for the rare use of iPSCs is that the control of differentiation into mature cell types is extremely complex, and in addition, immature cell phenotypes are al- ways present in cell culture (11,12). Regardless of the source and type of cells, we regard as simple in vitro mod- els the standard 2D single cell cultures (consisting of one type of cells grown in cell vessels) and their upgraded variants, such as 2D cocultures, 2D cultures grown in containers coated with extracellular matrix (ECM) components, and cultures grown on Transwell® plates. These are sim- ple 2D cell cocultures grown on plates that mimic more complex three-dimensional (3D) intercellular signalling (13,14). In fact, such 2D in vitro models have many drawbacks. In addition to the fact that cell vessels differ statistically significantly from the native tissue in structure, me- chanical properties, topography and com- position, these cells cannot communicate with each other auto- and paracrineally in all spatial dimensions. Also, cells are unevenly exposed to concentration gra- dients of oxygen, nutrients, and biologi- cally active molecules, leading to the fact that cells grown in 2D cultures do not show the correct morphology and often do not express the appropriate pheno- type long enough. Therefore, they do not Figure 1: Areas, where in vitro models are used. In vitro models Regenerative medicine Disease modelling Pharmacological and toxicological studies Personalized therapy Detection and testing of active substances Food testing and nutriceutics Tissue engineering Basic research 77 REVIEW ARTICLE In vitro models of the endocrine pancreas allow the performance of more complex and time-consuming experiments. As a result, simple in vitro models do not mim- ic the key characteristics and functions of the native tissue (15,16). In contrast to 2D cultures, cells grown in 3D cultures estab- lish complex interactions with the ECM and neighbouring cells in three spatial di- mensions, thus better mimicking the bio- chemistry and mechanics of the original microenvironment. Therefore, in order to build functionally more complex mod- els, in vitro cells must be grown in a 3D environment or on 3D cell scaffolds, i.e., bioscaffolds that mimic the original ECM in which cells grow, differentiate, prolifer- ate, and communicate in all spatial dimen- sions (17). 2.2 Advanced in vitro models Advanced in vitro models are defined as synthetic alternative experimental sys- tems based on living human cells that mimic the physiology of a tissue or organ in vitro using modern TE approaches and micro-additive manufacturing. Within the framework of this definition, the aim is not to perfectly mimic the complex archi- tecture and function of a tissue or organ, but rather the model reproduces, at least representatively, only the key functions that we want to mimic in vitro. Advanced 3D in vitro models are particularly useful when conventional 2D cell cultures do not replicate the dynamic responses of native tissues well enough (16,18). The basic ele- ments (Figure 2) that must be considered in the construction of the in vitro model are the source and type of cells (9), phys- icochemical stimuli (19) and biologically active molecules (biochemical stimuli) (20,21), which promote the desired cellu- lar phenotype. The simplest 3D in vitro models include spheroids and organoids grown in special cell vessels to which cells do not adhere. With appropriate biochemical stimuli, cells organize themselves into a simple spherical 3D shape. iPSCs are the most commonly used. As the name suggests, ex vivo organoids mimic the basic hierar- chical structure and physiology of organs, while spheroids do not have a clearly de- fined internal structure and cellular orga- nization (22). Organoids are mainly used for basic in vitro research dealing with or- ganogenesis and the course of the disease (23). In addition to the fact that organoids differ significantly in size and shape, the main limitation of both models is size, as cells within the 3D construct rapidly nec- rotize due to limited diffusion (24). To construct larger and more complex 3D constructs, it is extremely important to build cellular bioscaffolds with adequate porosity, facilitating the access of oxygen and nutrients to the cells (17,25). When constructing advanced 3D in vitro models, it is, therefore, necessary to additionally select appropriate build- ing blocks for the production of cellular scaffolds with appropriate structural and mechanical properties. The nano-, micro- and macro-properties of in vitro models should be adapted to mimic the charac- teristics of the native or diseased tissue. Emphasis should also be placed on mim- icking the mechanical conditions in which cells grow. Therefore, suitable biocompati- ble materials with appropriate mechanical properties must be selected. An important part of modelling also involves mimicking concentration gradients of nutrients, gas- es, pH, and metabolites (9). 3 Pancreas Ever since the development of the in vitro model, one of the primary goals has been to develop a functional in vitro mod- el of pancreas, and in particular the islets 78 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 of Langerhans, due to the desire to study the course of type 1 and 2 diabetes (lat. Diabetes mellitus, DM). The normal func- tioning of the human body depends on the precise regulation of blood glucose levels. The pancreas, more precisely the beta cells located in the islets of Langerhans, play a central role in maintaining the dynamic balance of glucose. These secrete insulin when the concentration of energy-rich molecules in the blood (especially glu- cose) and certain hormones (e.g. GIP and GLP-1 incretins) increases and under the influence of neurotransmitters (e.g. CCK and acetylcholine) (26,27). The pancreas is a retroperitoneal organ about 14-18 cm in size, consisting of three main anatomical parts: the head, body and tail. It is a gland that is structurally and functionally divid- ed into a larger (~ 95%), exocrine portion, consisting mainly of ductal and acinar cells, and a smaller (~ 5%), endocrine por- tion, consisting of islets of Langerhans. Acinar cells secrete digestive enzymes indirectly into the duodenum, which are responsible for breaking down proteins, lipids and carbohydrates. Ductal cells, on the other hand, secrete fluid rich in bicar- bonate anions (28). Endocrine tissues in humans are represented by about a million endocrine microorganisms, 50-500 µm in size, called islets of Langerhans. Each islet (even in different species) contains about 1,000 cells. The most numerous are beta cells (50-60%), which secrete insulin, fol- lowed by alpha cells (35-40%), which se- crete glucagon, and delta cells (10-15%), which secrete somatostatin. Less repre- sented are PP or gamma cells that secrete pancreatic polypeptide and epsilon cells that secrete ghrelin (29). DM types 1 and 2 are considered to be among the most important disorders in the functioning of the endocrine portion of the pancreas. Diabetes is a metabolic disease character- ized by the inability to regulate blood glu- cose homeostasis. In particular, (auto) im- mune-mediated type 1 diabetes is caused by an absolute lack of insulin due to the destruction of beta cells (30). DM type 2 is a complex polygenic disease that is also influenced by a number of environmen- tal and epigenetic factors. It is caused by a reduced sensitivity of the target tissues to insulin with initial compensatory in- crease but relative insufficiency of insulin secretion. This ultimately leads to secreto- ry dysfunction, beta cell failure, and abso- lute insulin deficiency (31-33). Both types of disease can lead to acute and chronic complications, including cardiovascular problems, renal failure, neurological dam- age, vision loss, and generally increased patient mortality (34,35). The most basic research on the origin and development of diabetes uses a number of different animal models (predominantly rodents, especial- ly mice) (36). Despite many similarities, there are many structural and physiolog- ical differences in the islets of Langerhans between humans and rodents, leading to differences in functional linkage between cells and finally to differences in the com- plex dynamics of insulin secretion (26,28). This means that the results obtained in animal models cannot always be reliably transferred to humans in all respects. 4 In vitro models of the pancreas The development of a functional hu- man model of an in vitro pancreas would mark a breakthrough in basic diabetes re- search and lead to the development of new therapeutical substances for the treatment of diabetes. At the same time, it would greatly reduce the need to use animal Figure 2: Key elements to consider when building advanced 3D in vitro models. After selecting the appropriate source and cell type, important biochemical and physicochemical stimuli must be mimicked in vivo which ensure the maintenance of the desired cellular phenotype. It is also essential that 3D bioscaffolds mimic the key physical and structural properties of the original ECM. Advanced in 3D in vitro models Commercially available cell lines Primary cell cultures Induced pluripotent stem cells Source and type of cells Biochemical stimuli Physical and structural properties of bioscaolds Physicochemical stimuli Growth factors Cytokines Hormones Steroids Peptides Geometry Porosity Topography Hardness High elastic properties Concentration gradients pH Temperature Charge 79 REVIEW ARTICLE In vitro models of the endocrine pancreas important disorders in the functioning of the endocrine portion of the pancreas. Diabetes is a metabolic disease character- ized by the inability to regulate blood glu- cose homeostasis. In particular, (auto) im- mune-mediated type 1 diabetes is caused by an absolute lack of insulin due to the destruction of beta cells (30). DM type 2 is a complex polygenic disease that is also influenced by a number of environmen- tal and epigenetic factors. It is caused by a reduced sensitivity of the target tissues to insulin with initial compensatory in- crease but relative insufficiency of insulin secretion. This ultimately leads to secreto- ry dysfunction, beta cell failure, and abso- lute insulin deficiency (31-33). Both types of disease can lead to acute and chronic complications, including cardiovascular problems, renal failure, neurological dam- age, vision loss, and generally increased patient mortality (34,35). The most basic research on the origin and development of diabetes uses a number of different animal models (predominantly rodents, especial- ly mice) (36). Despite many similarities, there are many structural and physiolog- ical differences in the islets of Langerhans between humans and rodents, leading to differences in functional linkage between cells and finally to differences in the com- plex dynamics of insulin secretion (26,28). This means that the results obtained in animal models cannot always be reliably transferred to humans in all respects. 4 In vitro models of the pancreas The development of a functional hu- man model of an in vitro pancreas would mark a breakthrough in basic diabetes re- search and lead to the development of new therapeutical substances for the treatment of diabetes. At the same time, it would greatly reduce the need to use animal Figure 2: Key elements to consider when building advanced 3D in vitro models. After selecting the appropriate source and cell type, important biochemical and physicochemical stimuli must be mimicked in vivo which ensure the maintenance of the desired cellular phenotype. It is also essential that 3D bioscaffolds mimic the key physical and structural properties of the original ECM. Advanced in 3D in vitro models Commercially available cell lines Primary cell cultures Induced pluripotent stem cells Source and type of cells Biochemical stimuli Physical and structural properties of bioscaolds Physicochemical stimuli Growth factors Cytokines Hormones Steroids Peptides Geometry Porosity Topography Hardness High elastic properties Concentration gradients pH Temperature Charge models. Despite many years of developing in vitro models, there are many problems remaining in the area of the pancreas. The first problem in the cultivation of in vitro pancreatic cells arises with the demanding process of isolation and purification of vi- able pancreatic islets and cells within the islet. Islet of Langerhans cells are no longer capable of auto- and paracrine communi- cation soon after isolation, when they lose homo- and heterotypic intercellular con- tacts. In addition, they lose critical ECM contacts and basement membrane con- tacts, which ultimately leads to reduced viability and loss of cell function. The additionally high metabolic requirements and size of the islets limit the availability and access of oxygen and nutrients to the cells inside the islet. Due to limited dif- fusion, necrotic cell death begins rapidly. All this contributes to the fact that current 2D in vitro pancreatic cell models do not mimic the critical dynamics of insulin se- cretion when stimulated with glucose (37). 2D substrates thus restrict the growth of pancreatic cells, prevent the formation of complex 3D in vivo morphology, and do not mimic ECM cell-type contacts that are critical for normal endocrine cell function. Even more in favour of the importance of the 3D environment for the differentia- tion, growth and functionality of the islets of Langerhans is the fact that recently a di- rect link between the morphology of the pancreatic islet and endocrine differentia- tion was confirmed. In the differentiation of endocrine progenitor cells, the alpha cells that develop first migrate to the out- side of the islet and form a mantle, while the beta cells that form later remain in the nucleus of the islet. Such temporal and spatial proportionality leads to the typical spherical 3D architecture of the human pancreatic islet (alpha cell mantle and be- ta cell nucleus), which is essential for the normal functioning of the islet (38). 80 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 4.1 3D Biomimetic scaffolds mimic the ECM of the native tissue In the most elementary sense, the ECM is a natural biologically active cellular scaffold composed of structural (colla- gen, laminins, fibronectins) and signalling proteins, polysaccharides, glycoproteins, proteoglycans, biologically active mole- cules, electrolytes, and water (39). In in vi- vo conditions, the ECM creates a complex 3D framework by providing mechanical support to cells and, in addition to inter- cellular communication in all spatial di- mensions, enables key cellular processes such as adhesion, migration, proliferation, and differentiation (40). ECM plays a key role in the microenvironment of pancre- atic cells (41), so it is extremely important to mimic the basic properties of the origi- nal ECM using 3D cellular scaffolds in the construction of an advanced in vitro mod- el of the pancreas (42,43). In general, cellular scaffolds must rep- licate the complex 3D microarchitecture, hierarchical structure, and ECM compo- sition of the native tissue to be mimicked in vitro. The basic building blocks, 3D structure, and composition of the pancre- atic ECM are best mimicked by scaffolds constructed of natural polymers (primari- ly polysaccharides). These form hydrogels and have a positive effect on the growth and viability of pancreatic cells (44). Therefore, to build advanced in vitro mod- els of the pancreas (islets of Langerhans), great effort is invested in the development of 3D biomimetic cellular scaffolds that mimic the basic building blocks of the original microenvironment (ECM) with which pancreatic cells are surrounded. A key criterion in the construction of the scaffold is the choice of the appropriate material. It must be based on the mechan- ical properties of the tissue, as the surface of the material plays an important role in directing the growth and development of cells, and it is also the central interface for intercellular interactions (45). The pan- creas is a nonlinear visco-elastic soft tissue with low shear modulus (46). Therefore, natural polymer hydrogels, which have a high water content and exhibit similar structural and mechanical properties as pancreatic ECM, are the best materials for building cellular scaffolds (47). In addition to biological compatibility, the basic prop- erties that must be taken into account in the construction of the bioscaffold are the structure and elementary building blocks of the scaffold. Topographic, material, and physical characteristics of all size classes (e.g. macro-, nano-, and microtopography, macro-, and microporosity) are structur- al stimuli that guide cell behaviour. The choice of biocompatible materials must be based on the fact that both the mi- cro- and macro-properties of the scaffolds mimic the characteristics of the native or pathologically altered pancreatic tissue. The surface properties of scaffolds (e.g. roughness), their mechanical properties, microstructures (e.g. pore size and shape), and other characteristics (e.g. swelling and biodegradability of scaffolds) significantly affect the growth and phenotype of the cell (48). Pancreatic cells grown in vitro on 3D cellular scaffolds were able to differentiate into physiologically appropriate tissue, and at the same time, their morphology differed significantly from cells grown in 2D cell cultures. Compared to 2D sub- strates, 3D scaffolds made of polysaccha- rides also promoted better adhesion, pro- liferation, and survival of cells (49-51). Although materials of natural ori- gin represent a good choice for the con- struction of 3D biomimetic scaffolds, they cannot mimic the complexity of the ECM of the native tissue down to the last detail. Recently developed decellularized 81 REVIEW ARTICLE In vitro models of the endocrine pancreas extracellular matrix (dECM) techniques promise the possibility of building dECMs cellular scaffolds that almost completely mimic the complexity of the native tissue. DECM cell scaffolds can be obtained from a variety of tissues by a decellularization method, which typically involves the lysis and removal of cells from the tissue by per- fusion with deionized water or detergents (52,53). Thus, only tissue-specific ECM remains after the process, which typical- ly involves macromolecules such as col- lagens, laminins, fibronectin, elastin, and other tissue-specific glycosaminoglycans, cytokines, and growth factors. Recently, a group of scientists demonstrated that such dECM-specific scaffolds also significant- ly affect the function of pancreatic cells. Using the decellularization method, they successfully prepared cellular scaffolds from the pancreas with a unique composi- tion, physical structure, and biological ac- tivity. iPSC cells grown on specific pancre- atic scaffolds spontaneously differentiated into cells, similar to pancreatic cells, which also began to express corresponding genes (54). DECM prepared from pancreatic tis- sue has also been successfully used for 3D printing of biologically mimetic cellular scaffolds. Such 3D printed dECM scaffolds promoted the differentiation of iPSCs against the pancreatic phenotype while maintaining the long-term viability and function of isolated islets of Langerhans (55). The biggest problem with the use of dECM remains the fact that during the de- cellularization process, the highly specific spatial arrangement of proteins and other molecules is disrupted. Therefore, the cur- rent goal in development remains to find a balance between the complete removal of cellular components and the preservation of small vessels (capillaries) and other tis- sue structures. Toxic effects were also ob- served on cells grown on dECM scaffolds, most likely due to residual detergents used during the process (56). Currently, the biggest limitation in the construction of 3D scaffolds that support the growth of pancreatic cells in the long run is the inadequate mechanical prop- erties of materials, especially hydrogels. Various crosslinking techniques (e.g. ionic crosslinking), inorganic/organic additives (cellulose fibres, various nanoparticles) (57,58), or other synthetic materials (poly- caprolactone) are used to improve the mechanical stability of hydrogels. These improve the mechanical properties of the scaffold or provide a suitable basic frame- work (57,59). An additional problem is the fact that it is extremely difficult to adjust the aforementioned key characteristics of scaffolds (porosity, topography, mechani- cal properties, rate of decay and water up- take, rheological properties of materials) to fully mimic the properties of the native tissue. The development of hydrogel for- mulations, on the basis of which it would be possible to produce long-term stable cellular scaffolds and to which the desired properties could be additionally arbitrari- ly adjusted (e.g. visco-elastic, mechanical, surface), would mean remarkable progress in the construction of advanced in vitro models (60). 5 3D printing for building in vitro models Despite all the technological advances in TE, the development of physiologically relevant tissues, especially the pancreas, remains difficult. Due to the limitation in the diffusion of oxygen and nutrients, the building of constructs larger than 200 µm is extremely problematic (25). Progress towards building larger tissue constructs is hampered by the fact that vascular functionality must be incorporated into the scaffold to ensure and improve the transfer of oxygen and nutrients to the 82 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 cells while promoting the removal of their waste products. Construction of 3D vas- cular networks within tissue constructs plays a key role in the long-term survival and maintenance of cell viability in in vitro models (61,62). To date, researchers have not been able to develop cellular scaffolds to build in vitro soft tissue models that demonstrate adequate resolution, struc- tural integrity, and required biocompat- ibility at the same time (63). Several TE approaches (64-67) have been developed to address these problems, but most are limited to inducing in vitro angiogenesis. In addition to the fact that such approach- es require long-term incubation for ves- sel formation as well as the use of costly growth factors (e.g. vascular endothelial growth factor, VEGF), their main draw- back is limited repeatability and the inabil- ity to spatially control the distribution of blood vessels within the construct, which often does not allow perfusion at all. In re- cent years, additive approaches such as 3D printing have been used more frequently to produce cellular scaffolds based on bio- compatible materials, advanced in vitro models, and tissue constructs with blood vessels included (68). In doing so, various additive production techniques are devel- oped and used simultaneously (e.g. litho- graphic techniques, i.e., ink-jet printing, microextrusion methods) to build 3D cel- lular scaffolds that better mimic the archi- tecture, biochemistry, and functionality of native tissues. Individual techniques show certain advantages and disadvantages (52). Among them, 3D bioprinting represents a new and most promising method used, which is expected to revolutionize the field of building advanced in vitro models. In addition to the ability to simultaneously use a wide range of biocompatible materi- als and exceptional application versatility (69,70), the 3D biological printer allows the loading of material with a micrometre spatial resolution (71) under cell-friendly conditions such as low shear forces (72,73). This gives the 3D printer a great advantage over other conventional techniques for the preparation of cellular scaffolds, which are often limited by the control of the 3D shape, the spatial arrangement of individ- ual components of the material and thus the local distribution of the density of ma- terial and cells (74). 5.1 Core/Shell 3D printing technique for building advanced in vitro models As already mentioned, for the develop- ment of larger and physiologically more complex models in vitro, it is extremely important to build cellular scaffolds that allow the smooth flow of cellular medium or even mimic the basic functionality of the vessel lumen. Probably the best and simplest approach to building a 3D print- ed in vitro flow model is to include a con- nected network of hollow channels inside the tissue scaffold. The construction of such a bioscaffold would reduce the like- lihood of the formation of necrotic areas in the construct and also solve many oth- er already mentioned shortcomings of the existing models (73). Endothelial cells can be further populated into the hollow ca- nals of the scaffolds, thus gaining the abil- ity to mimic synthetic vessels in 3D tissue models (75). In addition to the already mentioned facilitated diffusion of oxygen, nutrient inflow, and uninterrupted remov- al of CO2 and metabolites, such lumens of the channels can also fulfil other import- ant physiological tasks depending on the specifics of in vitro cultured tissue. In vitro endocrine tissue models allow secretion collection and assessment of the secreto- ry function of the construct, and exocrine tissues allow secretion and its collection for quantification. In order to build an 83 REVIEW ARTICLE In vitro models of the endocrine pancreas advanced 3D in vitro model of the entire pancreas, which would include both endo- crine and exocrine portions, in addition to the lumens where endocrine cells release their secretions, ducts for the release of enzymes must be additionally included in the biological scaffold, because their acti- vation could lead to autodigestion or even trigger in vitro pancreatitis of cultured tissue. Recently, a new version of the method of 3D printing of hollow channels (i.e., core/shell printing) (76-79) has attracted a lot of attention, as it promises fast, sim- ple, and repeatable construction of stable cellular scaffolds with built-in flow chan- nels. Using a coaxial nozzle allows so- called core/shell printing, 3D printing of two materials at the same time, one being extruded as a core filament (the core) and the other as a shell around it (the shell). The technique opens the possibility that by choosing biological materials with dif- ferent mechanical properties, the harder material structurally supports the softer material during and after printing. If to construct the scaffold a material is used that can be chemically crosslinked (e.g. al- ginate) and it is extruded as a shell while at the same time the crosslinking agent (e.g. CaCl2) is extruded into the core, then it is possible to construct a stable bios- caffold with hollow filaments in a single process step (80). Coaxial printing has already been used to build scaffolds with solid (81), so-called core/shell filaments (82), and hollow filaments (83). However, the materials used have not yet been op- timized for simultaneous cellular viabili- ty and even mechanical robustness of the constructs. Despite all the advantages of both ordinary 3D printing and so-called core/shell printing, the development of ap- propriate material formulations (ink) that simultaneously demonstrate all the nec- essary properties suitable for 3D printing and meet all biological requirements still presents a great challenge (84). 6 Tissue slice as a model of the pancreas in vitro Due to the many limitations and the complexity of building good 3D models of the pancreas, because this organ has a complex structure and function (28,85), isolated endocrine cells, especially beta cells, isolated acinar cells, isolated islets of Langerhans, and isolated ducts and ac- inuses are still mostly used for basic and translation studies of the pancreas (86,87). The results of these studies are often com- plemented by various in vivo measure- ments at the level of the whole organism (88). An important step towards the best possible 3D model for pancreatic tissue, which is alternative in many respects and partly complements the development of 3D models with the help of various media and 3D printing, has recently been devel- oped and the method of pancreatic tissue slices is increasingly being used (86,89). In this method, the pancreatic tis- sue of various types of model organisms (most often mice, possibly rats or pigs) or humans is cut into tissue slices about 100 micrometres thick. In contrast to the isolation of cells and islets, no enzymes are used, but only minimal mechanical stress due to cutting. Tissue slices contain intact islets of Langerhans, cut islets of Langerhans, large areas of intact acinus- es, and long sections of ducts of different orders of magnitude. One of the most im- portant properties of the tissue slices thus obtained is that the cells within the islets of Langerhans and within the acinuses are interconnected by various intercellular contacts while preserving the paracrine interaction within the endocrine portion, within the exocrine portion and between the two portions. To a greater extent than 84 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 in the isolation of islets and acinuses, the vessels, basement membrane, connective envelopes, immune cells, and other ele- ments of the mesenchyme are also pre- served, and thus also the 3D structure of the tissue (86,87,90,91). Acute pancreatic slice is thus a special form of primary cell culture that can be used for at least 24–48 hours without special additional scaffolds (87,92). In combination with electrophysio- logical measurements, intracellular cal- cium ion concentration measurements, secretion measurements, and various morphological measurements, the tissue slice method has been shown to be at least equivalent, in terms of results and repeat- ability, to cell and islet or acinus isolation methods (93-99). In many respects, how- ever, the tissue slice allows for more phys- iological data, especially when it comes to assessing communication between different cells (37,100-102). An addition- al important advantage of the tissue slice method is that it is well compatible with many different model organisms with flu- orescently labelled cells that interest us, and at the same time also with diabetes models that lead to the decay of most be- ta cells (e.g. streptozotocin model) and so they are not compatible with the isolation of cells or islets, as in these cases too little isolate is obtained (103,104). The results of morphological and functional measure- ments in the slice are becoming the gold standard in this field, as well as an import- ant reference for measurements in other 3D models. Finally, we should emphasize that tis- sue slices have recently been used in com- bination with special scaffolds that extend the lifespan of the ex vivo slice and its usefulness for studies by at least one week (87). When using human tissue, it is an- ticipated that scaffolds will need to allow tissue transfer for a duration of several days so that it can be used in studies in those parts of the world where there is no local source of human tissue or for mea- surements in specialized laboratories us- ing methods that are not available at the location of the source of human tissue. We are assuming that a larger or smaller part of the tissue from the slice can in the fu- ture also be used to build 3D models using the already mentioned advanced materials and techniques of combining cells with these materials (60). 7 A glance into the future Despite remarkable advances in both the broader interdisciplinary domain of tissue engineering and the narrower field of in vitro pancreas models, both fields still face many problems that need to be over- come. Currently, the most advanced 3D in vitro models mimic only the basic func- tions of individual tissues and organs, so despite the shortcomings, tissue slices are used as the gold standard for performing basic studies (especially with the pancre- as). A step in the right direction means the development of two seemingly unrelated fields of microfluidics and the production of computer microchips. The so-called organs-on-a-chip are cell cultures grown on advanced microfluidic devices (105). Microchip manufacturing techniques (e.g. soft lithography) (106) can produce high-precision flow scaffolds that better mimic physicochemical (concentration gradients of gases and nutrients), struc- tural (nano-topology), and biochemical stimuli (concentration gradients of bio- logically active molecules) of the tissues that are being mimicked. In addition, the cells in such devices are not grown in a static environment of cell vessels or 3D bioscaffolds, but are continuously exposed to the flow of cellular medium. Medium perfusion mimics key physicochemical 85 REVIEW ARTICLE In vitro models of the endocrine pancreas References 1. Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, et al.; Inflammation and Host Response to Injury, Large Scale Collaborative Research Program. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci USA. 2013;110(9):3507-12. DOI: 10.1073/ pnas.1222878110 PMID: 23401516 2. Langer R, Vacanti J. Advances in tissue engineering. J Pediatr Surg. 2016;51(1):8-12. DOI: 10.1016/j. jpedsurg.2015.10.022 PMID: 26711689 3. Atala A, Kasper FK, Mikos AG. Engineering complex tissues. Sci Transl Med. 2012;4(160):160rv12. DOI: 10.1126/scitranslmed.3004890 PMID: 23152327 4. Khademhosseini A, Vacanti JP, Langer R. Progress in tissue engineering. Sci Am. 2009;300(5):64-71. DOI: 10.1038/scientificamerican0509-64 PMID: 19438051 5. Wobma H, Vunjak-Novakovic G. Tissue Engineering and Regenerative Medicine 2015: A Year in Review. Tissue Eng Part B Rev. 2016;22(2):101-13. DOI: 10.1089/ten.teb.2015.0535 PMID: 26714410 6. Park KM, Shin YM, Kim K, Shin H. Tissue Engineering and Regenerative Medicine 2017: A Year in Review. Tissue Eng Part B Rev. 2018;24(5):327-44. DOI: 10.1089/ten.teb.2018.0027 PMID: 29652594 7. Skelin M, Rupnik M, Cencic A. Pancreatic beta cell lines and their applications in diabetes mellitus research. ALTEX. 2010;27(2):105-13. DOI: 10.14573/altex.2010.2.105 PMID: 20686743 8. Nestor CE, Ottaviano R, Reinhardt D, Cruickshanks HA, Mjoseng HK, McPherson RC, et al. Rapid reprogramming of epigenetic and transcriptional profiles in mammalian culture systems. Genome Biol. 2015;16(1):11. DOI: 10.1186/s13059-014-0576-y PMID: 25648825 stimuli (formation of shear forces and concentration gradients) to which cells are exposed in the source environment. At the same time, it facilitates the diffusion of oxygen and nutrients and enables the re- moval of waste products (107). The cells in the islet of Langerhans grown in such a microfluidic environment have shown, for example, morphology that is more correct and an improved viability compared to static cultures. In addition, when stimu- lated with glucose, beta cells maintained adequate insulin secretion dynamics for a longer time (108,109). Future research in the field of organs-on-a-chip will focus on the functional connection of individu- al organs and on the development of the so-called body-on-a-chip and additional coupling of the system with microsensors. This will not only provide insight into the functioning and responses of individu- al cells, but also into complex signalling and communication between different tissues and organs in real time. This will, among other things, open up a number of new possibilities in pharmacological and toxicological studies, and enable more advanced and targeted development of therapeutical substances, as well as the development of disease models affecting several organs at the same time (110). Current developments suggest that the si- multaneous development of (1) advanced materials with dynamically adaptable me- chanical, physicochemical, and structural properties, (2) more accurate micro-ad- ditive production methods to build more complex bioscaffolds (3), as well as the use of new cellular resources and (4) optimi- zation of cell culture conditions will lead to the construction of advanced in vitro models that will fully and representative- ly mimic the hierarchical structure and functionality of tissues or organs in vivo. 8 Acknowledgement The authors thank the Slovenian Research Agency for their financial as- sistance within the Young Researchers Programme and under contracts P-0036, P3-0396, I0-0029, N3-0048, N3-0133, J3- 9289 and J3-1762. 86 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 9. Horvath P, Aulner N, Bickle M, Davies AM, Nery ED, Ebner D, et al. Screening out irrelevant cell-based models of disease. Nat Rev Drug Discov. 2016;15(11):751-69. DOI: 10.1038/nrd.2016.175 PMID: 27616293 10. Singh VK, Kumar N, Kalsan M, Saini A, Chandra R. Mechanism of induction: induced pluripotent stem cells (iPSCs). J Stem Cells. 2015;10(1):43-62. PMID: 26665937 11. Wills QF, Boothe T, Asadi A, Ao Z, Warnock GL, Kieffer TJ, et al. Statistical approaches and software for clustering islet cell functional heterogeneity. Islets. 2016;8(2):48-56. DOI: 10.1080/19382014.2016.1150664 PMID: 26909740 12. de Lázaro I, Yilmazer A, Kostarelos K. Induced pluripotent stem (iPS) cells: a new source for cell-based therapeutics? J Control Release. 2014;185:37-44. DOI: 10.1016/j.jconrel.2014.04.011 PMID: 24746625 13. Elliott NT, Yuan F. A review of three-dimensional in vitro tissue models for drug discovery and transport studies. J Pharm Sci. 2011;100(1):59-74. DOI: 10.1002/jps.22257 PMID: 20533556 14. Breslin S, O’Driscoll L. Three-dimensional cell culture: the missing link in drug discovery. Drug Discov Today. 2013;18(5-6):240-9. DOI: 10.1016/j.drudis.2012.10.003 PMID: 23073387 15. Knight E, Przyborski S. Advances in 3D cell culture technologies enabling tissue-like structures to be created in vitro. J Anat. 2015;227(6):746-56. DOI: 10.1111/joa.12257 PMID: 25411113 16. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12(4):207-18. DOI: 10.1089/adt.2014.573 PMID: 24831787 17. Caddeo S, Boffito M, Sartori S. Tissue Engineering Approaches in the Design of Healthy and Pathological In Vitro Tissue Models. Front Bioeng Biotechnol. 2017;5:40. DOI: 10.3389/fbioe.2017.00040 PMID: 28798911 18. Mattei G, Giusti S, Ahluwalia A. Design criteria for generating physiologically relevant in vitro models in bioreactors. Processes (Basel). 2014;2(3):548-69. DOI: 10.3390/pr2030548 19. Di Nardo P, Minieri M, Ahluwalia A. Engineering the Stem Cell Niche and the Differentiative Micro-and Macroenvironment: Technologies and Tools for Applying Biochemical, Physical and Structural Stimuli and Their Effects on Stem Cells. In: Artmann GM, Minter S, Hescheler J, eds. Stem Cell Engineering. Berlin: Heidelberg: Springer; 2011. pp. 41-59. 20. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general approaches and a review of recent developments. J R Soc Interface. 2011;8(55):153-70. DOI: 10.1098/rsif.2010.0223 PMID: 20719768 21. Tayalia P, Mooney DJ. Controlled growth factor delivery for tissue engineering. Adv Mater. 2009;21(32- 33):3269-85. DOI: 10.1002/adma.200900241 PMID: 20882497 22. Huch M, Koo BK. Modeling mouse and human development using organoid cultures. Development. 2015;142(18):3113-25. DOI: 10.1242/dev.118570 PMID: 26395140 23. Clevers H. Modeling development and disease with organoids. Cell. 2016;165(7):1586-97. DOI: 10.1016/j. cell.2016.05.082 PMID: 27315476 24. Dutta D, Heo I, Clevers H. Disease Modeling in Stem Cell-Derived 3D Organoid Systems. Trends Mol Med. 2017;23(5):393-410. DOI: 10.1016/j.molmed.2017.02.007 PMID: 28341301 25. Lovett M, Lee K, Edwards A, Kaplan DL. Vascularization strategies for tissue engineering. Tissue Eng Part B Rev. 2009;15(3):353-70. DOI: 10.1089/ten.teb.2009.0085 PMID: 19496677 26. 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(6):109-39. DOI: 10.1080/19382014.2017.1342022 PMID: 28662366 27. Röder PV, Wu B, Liu Y, Han W. Pancreatic regulation of glucose homeostasis. Exp Mol Med. 2016;48(3):e219. DOI: 10.1038/emm.2016.6 PMID: 26964835 28. Dolenšek J, Rupnik MS, Stožer A. Structural similarities and differences between the human and the mouse pancreas. Islets. 2015;7(1):e1024405. DOI: 10.1080/19382014.2015.1024405 PMID: 26030186 29. Stožer A. Pancreas Physiology and Pathophysiology in Tissue Slices. Gastroenterolog. 2017;21(Suppl 3):68-73. 30. Tuomi T, Santoro N, Caprio S, Cai M, Weng J, Groop L. The many faces of diabetes: a disease with increasing heterogeneity. Lancet. 2014;383(9922):1084-94. DOI: 10.1016/S0140-6736(13)62219-9 PMID: 24315621 31. Stožer A, Hojs R, Dolenšek J. Beta Cell Functional Adaptation and Dysfunction in Insulin Resistance and the Role of Chronic Kidney Disease. Nephron. 2019;143(1):33-7. DOI: 10.1159/000495665 PMID: 30650405 32. Drong AW, Lindgren CM, McCarthy MI. The genetic and epigenetic basis of type 2 diabetes and obesity. Clin Pharmacol Ther. 2012;92(6):707-15. DOI: 10.1038/clpt.2012.149 PMID: 23047653 33. Gupta D, Krueger CB, Lastra G. Over-nutrition, obesity and insulin resistance in the development of β-cell dysfunction. Curr Diabetes Rev. 2012;8(2):76-83. DOI: 10.2174/157339912799424564 PMID: 22229253 34. King A, Bowe J. Animal models for diabetes: understanding the pathogenesis and finding new treatments. Biochem Pharmacol. 2016;99:1-10. DOI: 10.1016/j.bcp.2015.08.108 PMID: 26432954 87 REVIEW ARTICLE In vitro models of the endocrine pancreas 35. Harcourt BE, Penfold SA, Forbes JM. Coming full circle in diabetes mellitus: from complications to initiation. Nat Rev Endocrinol. 2013;9(2):113-23. DOI: 10.1038/nrendo.2012.236 PMID: 23296171 36. Rees DA, Alcolado JC. Animal models of diabetes mellitus. Diabet Med. 2005;22(4):359-70. DOI: 10.1111/j.1464-5491.2005.01499.x PMID: 15787657 37. Gosak M, Markovič R, Dolenšek J, Rupnik MS, Marhl M, Stožer A, et al. Network science of biological systems at different scales: a review. Phys Life Rev. 2018;24:118-35. DOI: 10.1016/j.plrev.2017.11.003 PMID: 29150402 38. Sharon N, Chawla R, Mueller J, Vanderhooft J, Whitehorn LJ, Rosenthal B, et al. A Peninsular Structure Coordinates Asynchronous Differentiation with Morphogenesis to Generate Pancreatic Islets. Cell. 2019;176(4):790-804.e13. DOI: 10.1016/j.cell.2018.12.003 PMID: 30661759 39. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014;15(12):786-801. DOI: 10.1038/nrm3904 PMID: 25415508 40. Petersen OW, Rønnov-Jessen L, Howlett AR, Bissell MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc Natl Acad Sci USA. 1992;89(19):9064-8. DOI: 10.1073/pnas.89.19.9064 PMID: 1384042 41. Ilieva A, Yuan S, Wang RN, Agapitos D, Hill DJ, Rosenberg L. Pancreatic islet cell survival following islet isolation: the role of cellular interactions in the pancreas. J Endocrinol. 1999;161(3):357-64. DOI: 10.1677/ joe.0.1610357 PMID: 10333538 42. Li W, Lee S, Ma M, Kim SM, Guye P, Pancoast JR, et al. Microbead-based biomimetic synthetic neighbors enhance survival and function of rat pancreatic β-cells. Sci Rep. 2013;3(1):2863. DOI: 10.1038/srep02863 PMID: 24091640 43. Mao GH, Chen GA, Bai HY, Song TR, Wang YX. The reversal of hyperglycaemia in diabetic mice using PLGA scaffolds seeded with islet-like cells derived from human embryonic stem cells. Biomaterials. 2009;30(9):1706-14. DOI: 10.1016/j.biomaterials.2008.12.030 PMID: 19135250 44. Nagata N, Gu Y, Hori H, Balamurugan AN, Touma M, Kawakami Y, et al. Evaluation of insulin secretion of isolated rat islets cultured in extracellular matrix. Cell Transplant. 2001;10(4-5):447-51. DOI: 10.3727/000000001783986549 PMID: 11549070 45. Maver T, Gradišnik L, Kurečič M, Hribernik S, Smrke DM, Maver U, et al. Layering of different materials to achieve optimal conditions for treatment of painful wounds. Int J Pharm. 2017;529(1-2):576-88. DOI: 10.1016/j.ijpharm.2017.07.043 PMID: 28723409 46. Wex C, Fröhlich M, Brandstädter K, Bruns C, Stoll A. Experimental analysis of the mechanical behavior of the viscoelastic porcine pancreas and preliminary case study on the human pancreas. J Mech Behav Biomed Mater. 2015;41:199-207. DOI: 10.1016/j.jmbbm.2014.10.013 PMID: 25460416 47. Tibbitt MW, Anseth KS. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol Bioeng. 2009;103(4):655-63. DOI: 10.1002/bit.22361 PMID: 19472329 48. Chang H-I, Wang Y. Cell responses to surface and architecture of tissue engineering scaffolds. London: IntechOpen Limited; 2011. DOI: 10.5772/21983 49. Tuch BE, Gao SY, Lees JG. Scaffolds for islets and stem cells differentiated into insulin-secreting cells. Front Biosci. 2014;19(1):126-38. DOI: 10.2741/4199 PMID: 24389176 50. Khorsandi L, Khodadadi A, Nejad-Dehbashi F, Saremy S. Three-dimensional differentiation of adipose- derived mesenchymal stem cells into insulin-producing cells. Cell Tissue Res. 2015;361(3):745-53. DOI: 10.1007/s00441-015-2140-9 PMID: 25795142 51. Aloysious N, Nair PD. Enhanced survival and function of islet-like clusters differentiated from adipose stem cells on a three-dimensional natural polymeric scaffold: an in vitro study. Tissue Eng Part A. 2014;20(9-10):1508-22. DOI: 10.1089/ten.tea.2012.0615 PMID: 24359126 52. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol. 2014;32(8):773-85. DOI: 10.1038/ nbt.2958 PMID: 25093879 53. Baptista PM, Orlando G, Mirmalek-Sani SH, Siddiqui M, Atala A, Soker S. Whole organ decellularization - a tool for bioscaffold fabrication and organ bioengineering. Annu Int Conf IEEE Eng Med Biol Soc. 2009;2009:6526-9. DOI: 10.1109/IEMBS.2009.5333145 PMID: 19964173 54. Berger C, Bjørlykke Y, Hahn L, Mühlemann M, Kress S, Walles H, et al. Matrix decoded - A pancreatic extracellular matrix with organ specific cues guiding human iPSC differentiation. Biomaterials. 2020;244:119766. DOI: 10.1016/j.biomaterials.2020.119766 PMID: 32199284 55. Kim J, Shim IK, Hwang DG, Lee YN, Kim M, Kim H, et al. 3D cell printing of islet-laden pancreatic tissue- derived extracellular matrix bioink constructs for enhancing pancreatic functions. J Mater Chem B Mater Biol Med. 2019;7(10):1773-81. DOI: 10.1039/C8TB02787K PMID: 32254919 56. Sullivan DC, Mirmalek-Sani SH, Deegan DB, Baptista PM, Aboushwareb T, Atala A, et al. Decellularization methods of porcine kidneys for whole organ engineering using a high-throughput system. Biomaterials. 2012;33(31):7756-64. DOI: 10.1016/j.biomaterials.2012.07.023 PMID: 22841923 88 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 57. Lee VK, Dai G. Printing of Three-Dimensional Tissue Analogs for Regenerative Medicine. Ann Biomed Eng. 2017;45(1):115-31. DOI: 10.1007/s10439-016-1613-7 PMID: 27066784 58. Utech S, Boccaccini AR. A review of hydrogel-based composites for biomedical applications: enhancement of hydrogel properties by addition of rigid inorganic fillers. J Mater Sci. 2016;51(1):271-310. DOI: 10.1007/s10853-015-9382-5 59. Do AV, Khorsand B, Geary SM, Salem AK. 3D Printing of Scaffolds for Tissue Regeneration Applications. Adv Healthc Mater. 2015;4(12):1742-62. DOI: 10.1002/adhm.201500168 PMID: 26097108 60. Milojević M, Gradišnik L, Stergar J, Skelin Klemen M, Stožer A, Vesenjak M, et al. Development of multifunctional 3D printed bioscaffolds from polysaccharides and NiCu nanoparticles and their application. Appl Surf Sci. 2019;488:836-52. DOI: 10.1016/j.apsusc.2019.05.283 61. Rouwkema J, Rivron NC, van Blitterswijk CA. Vascularization in tissue engineering. Trends Biotechnol. 2008;26(8):434-41. DOI: 10.1016/j.tibtech.2008.04.009 PMID: 18585808 62. Bae H, Puranik AS, Gauvin R, Edalat F, Carrillo-Conde B, Peppas NA, et al. Building vascular networks. Sci Transl Med. 2012;4(160). DOI: 10.1126/scitranslmed.3003688 PMID: 23152325 63. Štumberger G, Vihar B. Freeform Perfusable Microfluidics Embedded in Hydrogel Matrices. Materials (Basel). 2018;11(12):2529. DOI: 10.3390/ma11122529 PMID: 30545119 64. Ibrahim M, Richardson MK. Beyond organoids: in vitro vasculogenesis and angiogenesis using cells from mammals and zebrafish. Reprod Toxicol. 2017;73:292-311. DOI: 10.1016/j.reprotox.2017.07.002 PMID: 28697965 65. Sorrell JM, Baber MA, Caplan AI. Influence of adult mesenchymal stem cells on in vitro vascular formation. Tissue Eng Part A. 2009;15(7):1751-61. DOI: 10.1089/ten.tea.2008.0254 PMID: 19196139 66. Davies NH, Schmidt C, Bezuidenhout D, Zilla P. Sustaining neovascularization of a scaffold through staged release of vascular endothelial growth factor-A and platelet-derived growth factor-BB. Tissue Eng Part A. 2012;18(1-2):26-34. DOI: 10.1089/ten.tea.2011.0192 PMID: 21895488 67. Li X, He J, Zhang W, Jiang N, Li D. Additive manufacturing of biomedical constructs with biomimetic structural organizations. Materials (Basel). 2016;9(11):909. DOI: 10.3390/ma9110909 PMID: 28774030 68. Maver T, Smrke D, Kurečič M, Gradišnik L, Maver U, Kleinschek KS. Combining 3D printing and electrospinning for preparation of pain-relieving wound-dressing materials. J Sol-Gel Sci Technol. 2018;88(1):1-16. DOI: 10.1007/s10971-018-4630-1 69. Hinton TJ, Jallerat Q, Palchesko RN, Park JH, Grodzicki MS, Shue HJ, et al. Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels. Sci Adv. 2015;1(9):e1500758. DOI: 10.1126/sciadv.1500758 PMID: 26601312 70. Rocca M, Fragasso A, Liu W, Heinrich MA, Zhang YS. Embedded Multimaterial Extrusion Bioprinting. SLAS Technol. 2018;23(2):154-63. DOI: 10.1177/2472630317742071 PMID: 29132232 71. Derby B. Printing and prototyping of tissues and scaffolds. Science. 2012;338(6109):921-6. DOI: 10.1126/ science.1226340 PMID: 23161993 72. Kolesky DB, Truby RL, Gladman AS, Busbee TA, Homan KA, Lewis JA. 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Adv Mater. 2014;26(19):3124-30. DOI: 10.1002/ adma.201305506 PMID: 24550124 ; 2014. 73. Huang Y, Zhang XF, Gao G, Yonezawa T, Cui X. 3D bioprinting and the current applications in tissue engineering. Biotechnol J. 2017;12(8):1600734. DOI: 10.1002/biot.201600734 PMID: 28675678 74. Nakamura M, Iwanaga S, Henmi C, Arai K, Nishiyama Y. Biomatrices and biomaterials for future developments of bioprinting and biofabrication. Biofabrication. 2010;2(1):014110. DOI: 10.1088/1758- 5082/2/1/014110 PMID: 20811125 75. Hoch E, Tovar GE, Borchers K. Bioprinting of artificial blood vessels: current approaches towards a demanding goal. Eur J Cardiothorac Surg. 2014;46(5):767-78. DOI: 10.1093/ejcts/ezu242 PMID: 24970571 76. Yeo M, Lee JS, Chun W, Kim GH. An Innovative Collagen-Based Cell-Printing Method for Obtaining Human Adipose Stem Cell-Laden Structures Consisting of Core-Sheath Structures for Tissue Engineering. Biomacromolecules. 2016;17(4):1365-75. DOI: 10.1021/acs.biomac.5b01764 PMID: 26998966 77. Liu W, Zhong Z, Hu N, Zhou Y, Maggio L, Miri AK, et al. Coaxial extrusion bioprinting of 3D microfibrous constructs with cell-favorable gelatin methacryloyl microenvironments. Biofabrication. 2018;10(2):024102. DOI: 10.1088/1758-5090/aa9d44 PMID: 29176035 78. Gao Q, He Y, Fu JZ, Liu A, Ma L. Coaxial nozzle-assisted 3D bioprinting with built-in microchannels for nutrients delivery. Biomaterials. 2015;61:203-15. DOI: 10.1016/j.biomaterials.2015.05.031 PMID: 26004235 79. Akkineni AR, Ahlfeld T, Lode A, Gelinsky M. A versatile method for combining different biopolymers in a core/shell fashion by 3D plotting to achieve mechanically robust constructs. Biofabrication. 2016;8(4):045001. DOI: 10.1088/1758-5090/8/4/045001 PMID: 27716641 80. Milojević M, Vihar B, Banović L, Miško M, Gradišnik L, Zidarič T, et al. Core/shell Printing Scaffolds For Tissue Engineering Of Tubular Structures. J Vis Exp. 2019(151):e59951. DOI: 10.3791/59951 PMID: 31609306 89 REVIEW ARTICLE In vitro models of the endocrine pancreas 81. Colosi C, Shin SR, Manoharan V, Massa S, Costantini M, Barbetta A, et al. Microfluidic Bioprinting of Heterogeneous 3D Tissue Constructs Using Low-Viscosity Bioink. Adv Mater. 2016(28):677,84. DOI: 10.1002/adma.201503310 PMID: 26606883 82. Kim G, Ahn S, Kim Y, Cho Y, Chun W. Coaxial structured collagen–alginate scaffolds: fabrication, physical properties, and biomedical application for skin tissue regeneration. J Mater Chem. 2011;21(17):6165-72. DOI: 10.1039/c0jm03452e 83. Luo Y, Lode A, Gelinsky M. Direct plotting of three-dimensional hollow fiber scaffolds based on concentrated alginate pastes for tissue engineering. Adv Healthc Mater. 2013;2(6):777-83. DOI: 10.1002/ adhm.201200303 PMID: 23184455 84. Markstedt K, Mantas A, Tournier I, Martínez Ávila H, Hägg D, Gatenholm P. Martínez Ávila Hc, Hägg D, Gatenholm P. 3D bioprinting human chondrocytes with nanocellulose–alginate bioink for cartilage tissue engineering applications. Biomacromolecules. 2015;16(5):1489-96. DOI: 10.1021/acs.biomac.5b00188 PMID: 25806996 85. Dolenšek J, Pohorec V, Rupnik MS, Stožer A. Pancreas Physiology. In: Seicean A, ed. Challenges in Pancreatic Pathology. London: IntechOpen Limited; 20017. DOI: 10.5772/65895 86. Marciniak A, Cohrs CM, Tsata V, Chouinard JA, Selck C, Stertmann J, et al. Using pancreas tissue slices for in situ studies of islet of Langerhans and acinar cell biology. Nat Protoc. 2014;9(12):2809-22. DOI: 10.1038/ nprot.2014.195 PMID: 25393778 87. Marciniak A, Selck C, Friedrich B, Speier S. Mouse pancreas tissue slice culture facilitates long-term studies of exocrine and endocrine cell physiology in situ. PLoS One. 2013;8(11):e78706. DOI: 10.1371/journal. pone.0078706 PMID: 24223842 88. Speier S. Experimental approaches for high-resolution in vivo imaging of islet of Langerhans biology. Curr Diab Rep. 2011;11(5):420-5. DOI: 10.1007/s11892-011-0207-x PMID: 21701794 89. Speier S, Rupnik M. A novel approach to in situ characterization of pancreatic beta-cells. Pflugers Arch. 2003;446(5):553-8. DOI: 10.1007/s00424-003-1097-9 PMID: 12774232 90. Cohrs CM, Chen C, Jahn SR, Stertmann J, Chmelova H, Weitz J, et al. Vessel Network Architecture of Adult Human Islets Promotes Distinct Cell-Cell Interactions In Situ and Is Altered After Transplantation. Endocrinology. 2017;158(5):1373-85. DOI: 10.1210/en.2016-1184 PMID: 28324008 91. Weitz JR, Makhmutova M, Almaça J, Stertmann J, Aamodt K, Brissova M, et al. Mouse pancreatic islet macrophages use locally released ATP to monitor beta cell activity. Diabetologia. 2017. PMID: 28884198 92. Meneghel-Rozzo T, Rozzo A, Poppi L, Rupnik M. In vivo and in vitro development of mouse pancreatic beta-cells in organotypic slices. Cell Tissue Res. 2004;316(3):295-303. DOI: 10.1007/s00441-004-0886-6 PMID: 15085425 93. Dolenšek J, Stožer A, Skelin Klemen M, Miller EW, Slak Rupnik M. The relationship between membrane potential and calcium dynamics in glucose-stimulated beta cell syncytium in acute mouse pancreas tissue slices. PLoS One. 2013;8(12):e82374. DOI: 10.1371/journal.pone.0082374 PMID: 24324777 94. Stožer A, Dolenšek J, Rupnik MS. Glucose-stimulated calcium dynamics in islets of Langerhans in acute mouse pancreas tissue slices. PLoS One. 2013;8(1):e54638. DOI: 10.1371/journal.pone.0054638 PMID: 23358454 95. Dolenšek J, Špelič D, Klemen MS, Žalik B, Gosak M, Rupnik MS, et al. Membrane Potential and Calcium Dynamics in Beta Cells from Mouse Pancreas Tissue Slices: Theory, Experimentation, and Analysis. Sensors (Basel). 2015;15(11):27393-419. DOI: 10.3390/s151127393 PMID: 26516866 96. Skelin Klemen M, Dolenšek J, Stožer A, Rupnik M. Measuring Exocytosis in Endocrine Tissue Slices. In: Thorn P, ed. Exocytosis Methods. Humana Press; pp. 127-46. DOI: 10.1007/978-1-62703-676-4_7 97. Skelin M, Rupnik M. cAMP increases the sensitivity of exocytosis to Ca²+ primarily through protein kinase A in mouse pancreatic beta cells. Cell Calcium. 2011;49(2):89-99. DOI: 10.1016/j.ceca.2010.12.005 PMID: 21242000 98. Dolenšek J, Skelin M, Rupnik MS. Calcium dependencies of regulated exocytosis in different endocrine cells. Physiol Res. 2011;60:S29-38. DOI: 10.33549/physiolres.932176 PMID: 21777026 99. Stozer A, Dolensek J, Skelin M, Rupnik M. Cell physiology in tissue slices: studying beta cells in the islets of Langerhans = Celicna fiziologija v tkivnih rezinah: preucevanje celic beta v Langerhansovih otockih. Acta medico-biotechnica. 2013;6(1):20-32. 100. Markovič R, Stožer A, Gosak M, Dolenšek J, Marhl M, Rupnik MS. Progressive glucose stimulation of islet beta cells reveals a transition from segregated to integrated modular functional connectivity patterns. Sci Rep. 2015;5(1):7845. DOI: 10.1038/srep07845 PMID: 25598507 101. Gosak M, Stožer A, Markovič R, Dolenšek J, Perc M, Rupnik MS, et al. Critical and Supercritical Spatiotemporal Calcium Dynamics in Beta Cells. Front Physiol. 2017;8(1106):1106. DOI: 10.3389/ fphys.2017.01106 PMID: 29312008 102. Stožer A, Gosak M, Dolenšek J, Perc M, Marhl M, Rupnik MS, et al. Functional connectivity in islets of Langerhans from mouse pancreas tissue slices. PLOS Comput Biol. 2013;9(2):e1002923. DOI: 10.1371/ journal.pcbi.1002923 PMID: 23468610 90 METABOLIC AND HORMONAL DISORDERS Zdrav Vestn | January – February 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3001 103. Huang YC, Rupnik M, Gaisano HY. Unperturbed islet α-cell function examined in mouse pancreas tissue slices. J Physiol. 2011;589(Pt 2):395-408. DOI: 10.1113/jphysiol.2010.200345 PMID: 21078586 104. Huang YC, Rupnik MS, Karimian N, Herrera PL, Gilon P, Feng ZP, et al. In situ electrophysiological examination of pancreatic α cells in the streptozotocin-induced diabetes model, revealing the cellular basis of glucagon hypersecretion. Diabetes. 2013;62(2):519-30. DOI: 10.2337/db11-0786 PMID: 23043159 105. Low LA, Tagle DA. Tissue chips - innovative tools for drug development and disease modeling. Lab Chip. 2017;17(18):3026-36. DOI: 10.1039/C7LC00462A PMID: 28795174 106. Duffy DC, McDonald JC, Schueller OJ, Whitesides GM. Rapid prototyping of microfluidic systems in poly (dimethylsiloxane). Anal Chem. 1998;70(23):4974-84. DOI: 10.1021/ac980656z PMID: 21644679 107. Takebe T, Zhang B, Radisic M. Synergistic engineering: organoids meet organs-on-a-chip. Cell Stem Cell. 2017;21(3):297-300. DOI: 10.1016/j.stem.2017.08.016 PMID: 28886364 108. Sankar KS, Green BJ, Crocker AR, Verity JE, Altamentova SM, Rocheleau JV. Culturing pancreatic islets in microfluidic flow enhances morphology of the associated endothelial cells. PLoS One. 2011;6(9):e24904. DOI: 10.1371/journal.pone.0024904 PMID: 21961048 109. Silva PN, Green BJ, Altamentova SM, Rocheleau JV. A microfluidic device designed to induce media flow throughout pancreatic islets while limiting shear-induced damage. Lab Chip. 2013;13(22):4374-84. DOI: 10.1039/c3lc50680k PMID: 24056576 110. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol. 2014;32(8):760-72. DOI: 10.1038/ nbt.2989 PMID: 25093883