1
|
Peng X, Li L, Peng Y, Zhou G, An Z. Bioengineering and omics approaches for Type 1 diabetes practical research: advancements and constraints. Ann Med 2025; 57:2322047. [PMID: 39704022 DOI: 10.1080/07853890.2024.2322047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 12/21/2024] Open
Abstract
Insulin dependency arises from autoimmunity that targets the β cells of the pancreas, resulting in Type 1 diabetes (T1D). Despite the fact that T1D patients require insulin for survival, insulin does not provide a cure for this disease or prevent its complications. Despite extensive genetic, molecular, and cellular research on T1D over the years, the translation of this understanding into effective clinical therapies continues to pose a significant obstacle. It is therefore difficult to develop effective clinical treatment strategies without a thorough understanding of disease pathophysiology. Pancreatic tissue bioengineering models of human T1D offer a valuable approach to examining and controlling islet function while tackling various facets of the condition. And in recent years, due to advances in high-throughput omics analysis, the genotypic and molecular profiles of T1D have become finer tuned. The present article will examine recent progress in these areas, along with their utilization and constraints in the realm of T1D.
Collapse
Affiliation(s)
- Xi Peng
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yihua Peng
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guangju Zhou
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Thomson EA, Lee S, Xu H, Moeller H, Sands J, Lal RA, Annes JP, Poon ASY. Intermittent Low-Magnitude Pressure Applied Across Macroencapsulation Devices Enables Physiological Insulin Delivery Dynamics. Diabetes 2025; 74:873-884. [PMID: 40029687 DOI: 10.2337/db24-0818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Cadaveric islet and stem cell-derived transplantation hold promise as treatments for type 1 diabetes. To tackle the issue of immunocompatibility, numerous cellular macroencapsulation techniques that use diffusion to transport insulin across an immunoisolating barrier have been developed. However, despite several devices progressing to human clinical trials, none have successfully attained physiological glucose control or insulin independence. Based on empirical evidence, macroencapsulation methods with multilayered, high islet surface density are incompatible with on-demand insulin delivery and physiological glucose regulation when solely reliant on diffusion. An additional driving force is essential to overcome the distance limit of diffusion. In this study, we present both theoretical evidence and experimental validation that applying pressure, at levels comparable to physiological diastolic blood pressure, significantly enhances insulin flux across immunoisolation membranes, increasing it by nearly three orders of magnitude. This significant enhancement in transport rate allows for precise, subminute regulation of both bolus and basal insulin delivery. By incorporating this technique with a pump-based extravascular system, we demonstrate the ability to rapidly reduce glucose levels in diabetic rodent models, replicating the timescale and therapeutic effect of subcutaneous insulin injection or infusion. This advance provides a potential path toward achieving insulin independence with islet macroencapsulation. ARTICLE HIGHLIGHTS Numerous islet macroencapsulation techniques use diffusion to transport insulin across an immunoisolating barrier. Despite some devices reaching clinical trials, none have achieved physiological glucose control or insulin independence. Empirical evidence shows that high-density islet macroencapsulation methods cannot achieve on-demand insulin delivery and glucose regulation with diffusion alone. An additional driving force is needed. Appling a subminute pressure at physiological levels can achieve on-demand insulin delivery from macroencapsulated islets and glucose regulation. Incorporating pressure-based enhancements in macroencapsulation systems could lead to successful clinical outcomes.
Collapse
Affiliation(s)
- Ella A Thomson
- Department of Electrical Engineering, Stanford University, Stanford, CA
| | - Sooyeon Lee
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA
| | - Haixia Xu
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA
| | | | - Joanna Sands
- Department of Electrical Engineering, Stanford University, Stanford, CA
| | - Rayhan A Lal
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA
- Stanford Diabetes Research Center, Stanford, CA
- Division of Endocrinology, Department of Pediatrics, Stanford University, Stanford, CA
| | - Justin P Annes
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA
- Stanford Diabetes Research Center, Stanford, CA
- Safran ChEM-H, Stanford University, Stanford, CA
| | - Ada S Y Poon
- Department of Electrical Engineering, Stanford University, Stanford, CA
| |
Collapse
|
3
|
Shiwarski DJ, Hudson AR, Tashman JW, Bakirci E, Moss S, Coffin BD, Feinberg AW. 3D bioprinting of collagen-based high-resolution internally perfusable scaffolds for engineering fully biologic tissue systems. SCIENCE ADVANCES 2025; 11:eadu5905. [PMID: 40267204 PMCID: PMC12017336 DOI: 10.1126/sciadv.adu5905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025]
Abstract
Organ-on-a-chip and microfluidic systems have improved the translational relevance of in vitro systems; however, current manufacturing approaches impart limitations on materials selection, non-native mechanical properties, geometric complexity, and cell-driven remodeling into functional tissues. Here, we three-dimensionally (3D) bioprint extracellular matrix (ECM) and cells into collagen-based high-resolution internally perfusable scaffolds (CHIPS) that integrate with a vascular and perfusion organ-on-a-chip reactor (VAPOR) to form a complete tissue engineering platform. We improve the fidelity of freeform reversible embedding of suspended hydrogels (FRESH) bioprinting to produce a range of CHIPS designs fabricated in a one-step process. CHIPS exhibit size-dependent permeability of perfused molecules into the surrounding scaffold to support cell viability and migration. Lastly, we implemented multi-material bioprinting to control 3D spatial patterning, ECM composition, cellularization, and material properties to create a glucose-responsive, insulin-secreting pancreatic-like CHIPS with vascular endothelial cadherin+ vascular-like networks. Together, CHIPS and VAPOR form a platform technology toward engineering full organ-scale function for disease modeling and cell replacement therapy.
Collapse
Affiliation(s)
- Daniel J. Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Andrew R. Hudson
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Joshua W. Tashman
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Samuel Moss
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Brian D. Coffin
- Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Adam W. Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
4
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
5
|
Lallouet M, Olçomendy L, Gaitan J, Montiège K, Monchablon M, Pirog A, Chapeau D, Puginier E, Renaud S, Raoux M, Lang J. A microfluidic twin islets-on-chip device for on-line electrophysiological monitoring. LAB ON A CHIP 2025; 25:1831-1841. [PMID: 40042033 DOI: 10.1039/d4lc00967c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Pancreatic islets play a major role in glucose homeostasis as well as in diabetes, and islets-on-chip devices have been mainly developed using optical means for on-line monitoring. In contrast, no well-characterized electrophysiological platform for on-line analysis with unrivalled temporal resolution has been reported. Extracellular electrophysiology monitors two crucial parameters, islet β-cell activity and β-to-β-cell coupling, does not require chemical or genetic probes with inherent potential bias, is non-invasive and permits repetitive long-term monitoring. We have now developed and characterized a microfluidic islets-on-chip for combined electrophysiology (on-line) and hormone monitoring (off-line) with two chambers for concomitant monitoring. Fabrication of the device, based on commercial or easily manufacturable components, is within the reach of non-specialized laboratories. The chip permits convenient loading as well as long-term culture with comparable glucose kinetics and low shear stress in both chambers. An optimized flow rate did not alter islet β-cell electrical activity or coupling in response to glucose. Culturing for up to 8 days did not change islet survival as well as glucose-induced electrical or secretory kinetics of islet β-cells. The addition of a physiological amino acid mix, in the presence of elevated glucose, made a considerable change in the functional organisation of islet β-cell activity in terms of frequency and coupling, which explains the ensuing strong increase in insulin secretion. This device thus allows reliable long-term multiparametric on-line monitoring in two islet populations. The ease of fabrication, assembly and handling should permit widespread long-term on-line monitoring of islet activity in native micro-organs (e.g. controls/mutants), pseudo-islets or stem-cell-derived islet-like organoids.
Collapse
Affiliation(s)
- Marie Lallouet
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Loic Olçomendy
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Julien Gaitan
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Killian Montiège
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Marie Monchablon
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Antoine Pirog
- Junia, Electronics-Physics-Acoustics Department, F-59000 Lille, France
| | - Dorian Chapeau
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Emilie Puginier
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Sylvie Renaud
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Matthieu Raoux
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Jochen Lang
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| |
Collapse
|
6
|
Li K, Du S, Li H, Li Z, Zhu Q, Peng Q, Liao B, Qi L. A novel three-dimensional co-culture model for studying exosome-mediated cell interactions in glioblastoma. Biochim Biophys Acta Gen Subj 2025; 1869:130752. [PMID: 39793675 DOI: 10.1016/j.bbagen.2024.130752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025]
Abstract
Three-dimensional(3D) cell culture systems provide a larger space for cell proliferation, which is crucial for simulating cellular behavior and drug responses in the tumor microenvironment. In this study, we developed a novel 3D co-culture system for cell interactions, utilizing a commercialized bioreactor-microcarrier system. Mesenchymal stem cells (MSCs) were extracted via enzymatic digestion, and markers CD105 and CD31 were identified. Cell growth was observed using AO and immunofluorescence staining. No significant differences in Ki67 and GFAP expression were found between 2D and 3D cultures, though the 3D system offered more space for proliferation and reduced contact inhibition. Therefore, this 3D culture system may represent the tumor microenvironment more accurately than 2D cultures and will facilitate the investigation of the characteristics and functions of exosomes derived from this system. Exosomes are nanoscale vesicles that mediate intercellular communication by transferring molecules such as miRNAs between cells. Exosomes from 3D cultures were collected via ultra-high-speed centrifugation and characterized using nano-flow cytometry, transmission electron microscopy, and western blotting for markers CD9, Alix, and TSG101. PKH26 staining revealed peak exosome uptake by tumor cells at 24 h and complete metabolism by 72 h. Exosomes from 3D cultures inhibited GBM cell proliferation, migration, and invasion. Lastly, miRNA sequencing of exosomes was performed. This study emphasizes the importance of creating 3D co-culture systems to advance cancer research and offers a helpful tool for studying the complex cell interaction environment of GBM and other malignancies.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China
| | - Siyuan Du
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China
| | - Haichao Li
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China
| | - Zhaohui Li
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China
| | - Qihui Zhu
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China
| | - Qian Peng
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China
| | - Baojian Liao
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China.
| | - Ling Qi
- Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, PR China.
| |
Collapse
|
7
|
Rajalekshmi R, Agrawal DK. Synergistic potential of stem cells and microfluidics in regenerative medicine. Mol Cell Biochem 2025; 480:1481-1493. [PMID: 39285093 PMCID: PMC11842489 DOI: 10.1007/s11010-024-05108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/27/2024] [Indexed: 02/21/2025]
Abstract
Regenerative medicine has immense potential to revolutionize healthcare by using regenerative capabilities of stem cells. Microfluidics, a cutting-edge technology, offers precise control over cellular microenvironments. The integration of these two fields provides a deep understanding of stem cell behavior and enables the development of advanced therapeutic strategies. This critical review explores the use of microfluidic systems to culture and differentiate stem cells with precision. We examined the use of microfluidic platforms for controlled nutrient supply, mechanical stimuli, and real-time monitoring, providing an unprecedented level of detail in studying cellular responses. The convergence of stem cells and microfluidics holds immense promise for tissue repair, regeneration, and personalized medicine. It offers a unique opportunity to revolutionize the approach to regenerative medicine, facilitating the development of advanced therapeutic strategies and enhancing healthcare outcomes.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
8
|
Kim M, Cho S, Hwang DG, Shim IK, Kim SC, Jang J, Jang J. Bioprinting of bespoke islet-specific niches to promote maturation of stem cell-derived islets. Nat Commun 2025; 16:1430. [PMID: 39920133 PMCID: PMC11805982 DOI: 10.1038/s41467-025-56665-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Pancreatic islets are densely packed cellular aggregates containing various hormonal cell types essential for blood glucose regulation. Interactions among these cells markedly affect the glucoregulatory functions of islets along with the surrounding niche and pancreatic tissue-specific geometrical organization. However, stem cell (SC)-derived islets generated in vitro often lack the three-dimensional extracellular microenvironment and peri-vasculature, which leads to the immaturity of SC-derived islets, reducing their ability to detect glucose fluctuations and insulin release. Here, we bioengineer the in vivo-like pancreatic niches by optimizing the combination of pancreatic tissue-specific extracellular matrix and basement membrane proteins and utilizing bioprinting-based geometrical guidance to recreate the spatial pattern of islet peripheries. The bioprinted islet-specific niche promotes coordinated interactions between islets and vasculature, supporting structural and functional features resembling native islets. Our strategy not only improves SC-derived islet functionality but also offers significant potential for advancing research on islet development, maturation, and diabetic disease modeling, with future implications for translational applications.
Collapse
Affiliation(s)
- Myungji Kim
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Seungyeun Cho
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dong Gyu Hwang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - In Kyong Shim
- Asan Institute for Life Science, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Song Cheol Kim
- Asan Institute for Life Science, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Jiwon Jang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinah Jang
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Moss SP, Bakirci E, Feinberg AW. Engineering the 3D structure of organoids. Stem Cell Reports 2025; 20:102379. [PMID: 39706178 PMCID: PMC11784486 DOI: 10.1016/j.stemcr.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024] Open
Abstract
Organoids form through the sel f-organizing capabilities of stem cells to produce a variety of differentiated cell and tissue types. Most organoid models, however, are limited in terms of the structure and function of the tissues that form, in part because it is difficult to regulate the cell type, arrangement, and cell-cell/cell-matrix interactions within these systems. In this article, we will discuss the engineering approaches to generate more complex organoids with improved function and translational relevance, as well as their advantages and disadvantages. Additionally, we will explore how biofabrication strategies can manipulate the cell composition, 3D organization, and scale-up of organoids, thus improving their utility for disease modeling, drug screening, and regenerative medicine applications.
Collapse
Affiliation(s)
- Samuel P Moss
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Suryana M, Produit T, Yang H, Birarda G, Shanmugar JV, Krivitsky L, Paterova A, Grenci G. Infrared imaging with visible light in microfluidic devices: the water absorption barrier. Analyst 2025; 150:405-413. [PMID: 39692693 DOI: 10.1039/d4an01201a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Infrared spectro-microscopy is a powerful technique for analysing chemical maps of cells and tissues for biomedical and clinical applications, yet the strong water absorption in the mid-infrared region is a challenge to overcome, as it overlaps with the spectral fingerprints of biological components. Microfluidic chips offer ultimate control over the water layer thickness and are increasingly used in infrared spectro-microscopy. However, the actual impact of the water layer thickness on the instrument's performance is often left to the experimentalist's intuition and the peculiarities of specific instruments. Aiming to experimentally test the amount of absorption introduced by water with varying layer thicknesses, we fabricated a set of microfluidic devices with three controlled chamber thicknesses, each comprising a simple test pattern made of a well-known photoresist SU-8. We employed two infrared spectro-microscopy methods for measurements. The first method involves using a standard FTIR microscope with a benchtop infrared light source. The second method is a quantum infrared microscopy technique, where infrared imaging is achieved by detecting correlated photons in the visible range. We demonstrated that both methods enable the measurement of the absorption spectrum in the mid-IR region, even in the presence of up to a 30 μm thick water layer on top of a sample pattern. Additionally, the Q-IR technique offers practical advantages over synchrotron-based FTIR, such as reduced complexity, cost, and ease of operation.
Collapse
Affiliation(s)
- Mona Suryana
- Mechanobiology Institute (MBI), National University of Singapore, 5A Engineering Drive 1, 117411, Republic of Singapore.
| | - Thomas Produit
- A*STAR Quantum Innovation Centre (Q.InC), Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634, Republic of Singapore
| | - Hongzhi Yang
- A*STAR Quantum Innovation Centre (Q.InC), Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634, Republic of Singapore
| | - Giovanni Birarda
- SISSI Beamline, Elettra Synchrotron Light Facility, Basovizza (Ts, IT), Italy
| | - Jegan Vishnuwardhana Shanmugar
- Mechanobiology Institute (MBI), National University of Singapore, 5A Engineering Drive 1, 117411, Republic of Singapore.
| | - Leonid Krivitsky
- A*STAR Quantum Innovation Centre (Q.InC), Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634, Republic of Singapore
| | - Anna Paterova
- A*STAR Quantum Innovation Centre (Q.InC), Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634, Republic of Singapore
| | - Gianluca Grenci
- Mechanobiology Institute (MBI), National University of Singapore, 5A Engineering Drive 1, 117411, Republic of Singapore.
- Biomedical Engineering Department, National University of Singapore, 4 Engineering Drive 3 Block 4, Republic of Singapore 117583
| |
Collapse
|
11
|
Moeun BN, Lemaire F, Smink AM, Ebrahimi Orimi H, Leask RL, de Vos P, Hoesli CA. Oxygenation and function of endocrine bioartificial pancreatic tissue constructs under flow for preclinical optimization. J Tissue Eng 2025; 16:20417314241284826. [PMID: 39866963 PMCID: PMC11758540 DOI: 10.1177/20417314241284826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/02/2024] [Indexed: 01/28/2025] Open
Abstract
Islet transplantation and more recently stem cell-derived islets were shown to successfully re-establish glycemic control in people with type 1 diabetes under immunosuppression. These results were achieved through intraportal infusion which leads to early graft losses and limits the capacity to contain and retrieve implanted cells in case of adverse events. Extra-hepatic sites and encapsulation devices have been developed to address these challenges and potentially create an immunoprotective or immune-privileged environment. Many strategies have achieved reversal of hyperglycemia in diabetic rodents. So far, the results have been less promising when transitioning to humans and larger animal models due to challenges in oxygenation and insulin delivery. We propose a versatile in vitro perfusion system to culture and experimentally study the function of centimeter-scale tissues and devices for insulin-secreting cell delivery. The system accommodates various tissue geometries, experimental readouts, and oxygenation tensions reflective of potential transplantation sites. We highlight the system's applications by using case studies to explore three prominent bioartificial endocrine pancreas (BAP) configurations: (I) with internal flow, (II) with internal flow and microvascularized, and (III) without internal flow. Oxygen concentration profiles modeled computationally were analogous to viability gradients observed experimentally through live/dead endpoint measurements and in case I, time-lapse fluorescence imaging was used to monitor the viability of GFP-expressing cells in real time. Intervascular BAPs were cultured under flow for up to 3 days and BAPs without internal flow for up to 7 days, showing glucose-responsive insulin secretion quantified through at-line non-disruptive sampling. This system can complement other preclinical platforms to de-risk and optimize BAPs and other artificial tissue designs prior to clinical studies.
Collapse
Affiliation(s)
- Brenden N Moeun
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
| | - Florent Lemaire
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Richard L Leask
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
12
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
13
|
Shi J, Li J, Su W, Xue C, Zhang Y, Gao X. Engineered microenvironments and pancreatic islet-on-chips for screening sugar substitute and antidiabetic compounds. Food Res Int 2024; 196:115084. [PMID: 39614569 DOI: 10.1016/j.foodres.2024.115084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 12/01/2024]
Abstract
Recent advancements in the food industry have rekindled interest in the safety of food additives, such as sugar substitutes and food pigments. Consequently, the main purpose of this study was to develop models that can more accurately predict the effects of these additives on the human body. In response to this demand, we have created an innovative pancreas islet-on-a-chip system featuring a concentration gradient generator and a perfusable 3D cell culture array. This setup facilitates the 3D culture of pseudo-islets under stable biochemical and biophysical conditions. When compared to static culture environments, our dynamic environment maintains islet cell viability at over 95 %, resulting in larger cell clusters that exhibit a higher tendency for aggregation up to 30 μm. Furthermore, the expression levels of key factors integral to islet development, namely INS-1, INS-2, and PDX-1, increased by 4.5-fold, 1.9-fold, and 5.8-fold respectively in the dynamic environment. Utilizing this sophisticated pancreas islet-on-a-chip model, we discovered that the consumption of sugar substitutes like erythritol and sucralose for 1 h does not impact insulin secretion levels. In contrast, the administration of glucagon-like peptide 1 (GLP-1), GLP-1 receptor (GLP-1R) agonist exendin-4, curcumin, and a combination therapy group led to a substantial increase in insulin secretion levels (p < 0.01). Such engineered microenvironments and pancreatic islet-on-chips offer a groundbreaking platform for evaluating sugar substitutes and antidiabetic compounds.
Collapse
Affiliation(s)
- Jingyan Shi
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Jianing Li
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Wentao Su
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Chang Xue
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Yuan Zhang
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
14
|
Kim D, Lim H, Youn J, Park TE, Kim DS. Scalable production of uniform and mature organoids in a 3D geometrically-engineered permeable membrane. Nat Commun 2024; 15:9420. [PMID: 39482314 PMCID: PMC11528013 DOI: 10.1038/s41467-024-53073-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
The application of organoids has been limited by the lack of methods for producing uniformly mature organoids at scale. This study introduces an organoid culture platform, called UniMat, which addresses the challenges of uniformity and maturity simultaneously. UniMat is designed to not only ensure consistent organoid growth but also facilitate an unrestricted supply of soluble factors by a 3D geometrically-engineered, permeable membrane-based platform. Using UniMat, we demonstrate the scalable generation of kidney organoids with enhanced uniformity in both structure and function compared to conventional methods. Notably, kidney organoids within UniMat show improved maturation, showing increased expression of nephron transcripts, more in vivo-like cell-type balance, enhanced vascularization, and better long-term stability. Moreover, UniMat's design offers a more standardized organoid model for disease modeling and drug testing, as demonstrated by polycystic-kidney disease and acute kidney injury modeling. In essence, UniMat presents a valuable platform for organoid technology, with potential applications in organ development, disease modeling, and drug screening.
Collapse
Affiliation(s)
- Dohui Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Hyeonji Lim
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea.
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea.
| |
Collapse
|
15
|
Aleman J, K R, Wiegand C, Schurdak ME, Vernetti L, Gavlock D, Reese C, DeBiasio R, LaRocca G, Angarita YD, Gough A, Soto-Gutierrez A, Behari J, Yechoor VK, Miedel MT, Stern AM, Banerjee I, Taylor DL. A metabolic dysfunction-associated steatotic liver acinus biomimetic induces pancreatic islet dysfunction in a coupled microphysiology system. Commun Biol 2024; 7:1317. [PMID: 39397070 PMCID: PMC11471816 DOI: 10.1038/s42003-024-07006-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024] Open
Abstract
Preclinical and clinical studies suggest that lipid-induced hepatic insulin resistance is a primary defect that predisposes to dysfunction in islets, implicating a perturbed liver-pancreas axis underlying the comorbidity of T2DM and MASLD. To investigate this hypothesis, we developed a human biomimetic microphysiological system (MPS) coupling our vascularized liver acinus MPS (vLAMPS) with pancreatic islet MPS (PANIS) enabling MASLD progression and islet dysfunction to be assessed. The modular design of this system (vLAMPS-PANIS) allows intra-organ and inter-organ dysregulation to be deconvoluted. When compared to normal fasting (NF) conditions, under early metabolic syndrome (EMS) conditions, the standalone vLAMPS exhibited characteristics of early stage MASLD, while no significant differences were observed in the standalone PANIS. In contrast, with EMS, the coupled vLAMPS-PANIS exhibited a perturbed islet-specific secretome and a significantly dysregulated glucose stimulated insulin secretion response implicating direct signaling from the dysregulated liver acinus to the islets. Correlations between several pairs of a vLAMPS-derived and a PANIS-derived factors were significantly altered under EMS, as compared to NF conditions, mechanistically connecting MASLD and T2DM associated hepatic-factors with islet-derived GLP-1 synthesis and regulation. Since vLAMPS-PANIS is compatible with patient-specific iPSCs, this platform represents an important step towards addressing patient heterogeneity, identifying disease mechanisms, and advancing precision medicine.
Collapse
Affiliation(s)
- Julio Aleman
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Bioengineering, Pittsburgh, USA
| | - Ravikumar K
- University of Pittsburgh Department of Chemical and Petroleum Engineering, Pittsburgh, USA
| | - Connor Wiegand
- University of Pittsburgh Department of Chemical and Petroleum Engineering, Pittsburgh, USA
| | - Mark E Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
| | - Dillon Gavlock
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | - Celeste Reese
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | - Richard DeBiasio
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | - Greg LaRocca
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | | | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA
| | - Alejandro Soto-Gutierrez
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
- University of Pittsburgh Department of Pathology, Pittsburgh, USA
| | - Jaideep Behari
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Vijay K Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, USA
| | - Mark T Miedel
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
- University of Pittsburgh Department of Pathology, Pittsburgh, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA.
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA.
| | - Ipsita Banerjee
- University of Pittsburgh Department of Bioengineering, Pittsburgh, USA.
- University of Pittsburgh Department of Chemical and Petroleum Engineering, Pittsburgh, USA.
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA.
- University of Pittsburgh Department of Bioengineering, Pittsburgh, USA.
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA.
- University of Pittsburgh Liver Research Center, Pittsburgh, USA.
| |
Collapse
|
16
|
Mehta V, Karnam G, Madgula V. Liver-on-chips for drug discovery and development. Mater Today Bio 2024; 27:101143. [PMID: 39070097 PMCID: PMC11279310 DOI: 10.1016/j.mtbio.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Recent FDA modernization act 2.0 has led to increasing industrial R&D investment in advanced in vitro 3D models such as organoids, spheroids, organ-on-chips, 3D bioprinting, and in silico approaches. Liver-related advanced in vitro models remain the prime area of interest, as liver plays a central role in drug clearance of compounds. Growing evidence indicates the importance of recapitulating the overall liver microenvironment to enhance hepatocyte maturity and culture longevity using liver-on-chips (LoC) in vitro. Hence, pharmaceutical industries have started exploring LoC assays in the two of the most challenging areas: accurate in vitro-in vivo extrapolation (IVIVE) of hepatic drug clearance and drug-induced liver injury. We examine the joint efforts of commercial chip manufacturers and pharmaceutical companies to present an up-to-date overview of the adoption of LoC technology in the drug discovery. Further, several roadblocks are identified to the rapid adoption of LoC assays in the current drug development framework. Finally, we discuss some of the underexplored application areas of LoC models, where conventional 2D hepatic models are deemed unsuitable. These include clearance prediction of metabolically stable compounds, immune-mediated drug-induced liver injury (DILI) predictions, bioavailability prediction with gut-liver systems, hepatic clearance prediction of drugs given during pregnancy, and dose adjustment studies in disease conditions. We conclude the review by discussing the importance of PBPK modeling with LoC, digital twins, and AI/ML integration with LoC.
Collapse
Affiliation(s)
- Viraj Mehta
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Guruswamy Karnam
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Vamsi Madgula
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| |
Collapse
|
17
|
Li J, Sun L, Bian F, Pandol SJ, Li L. Emerging approaches for the development of artificial islets. SMART MEDICINE 2024; 3:e20230042. [PMID: 39188698 PMCID: PMC11235711 DOI: 10.1002/smmd.20230042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/05/2024] [Indexed: 08/28/2024]
Abstract
The islet of Langerhans, functioning as a "mini organ", plays a vital role in regulating endocrine activities due to its intricate structure. Dysfunction in these islets is closely associated with the development of diabetes mellitus (DM). To offer valuable insights for DM research and treatment, various approaches have been proposed to create artificial islets or islet organoids with high similarity to natural islets, under the collaborative effort of biologists, clinical physicians, and biomedical engineers. This review investigates the design and fabrication of artificial islets considering both biological and tissue engineering aspects. It begins by examining the natural structures and functions of native islets and proceeds to analyze the protocols for generating islets from stem cells. The review also outlines various techniques used in crafting artificial islets, with a specific focus on hydrogel-based ones. Additionally, it provides a concise overview of the materials and devices employed in the clinical applications of artificial islets. Throughout, the primary goal is to develop artificial islets, thereby bridging the realms of developmental biology, clinical medicine, and tissue engineering.
Collapse
Affiliation(s)
- Jingbo Li
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| | - Lingyu Sun
- Department of Clinical LaboratoryNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Feika Bian
- Department of Clinical LaboratoryNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Stephen J. Pandol
- Division of GastroenterologyDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Ling Li
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
18
|
Aleman J, Ravikumar K, Wiegand C, Schurdak ME, Vernetti L, Gavlock D, Reese C, DeBiasio R, LaRocca G, Angarita YD, Gough A, Soto-Gutierrez A, Behari J, Yechoor V, Miedel MT, Stern AM, Banerjee I, Taylor DL. A metabolic-dysfunction associated steatotic liver acinus biomimetic induces pancreatic islet dysfunction in a coupled microphysiology system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590598. [PMID: 38712135 PMCID: PMC11071380 DOI: 10.1101/2024.04.22.590598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Preclinical and clinical studies suggest that lipid-induced hepatic insulin resistance is a primary defect that predisposes to dysfunction in pancreatic islets, implicating a perturbed liver-pancreas axis underlying the comorbidity of T2DM and MASLD. To investigate this hypothesis, we developed a human biomimetic microphysiological system (MPS) coupling our vascularized liver acinus MPS (vLAMPS) with primary islets on a chip (PANIS) enabling MASLD progression and islet dysfunction to be quantitatively assessed. The modular design of this system (vLAMPS-PANIS) allows intra-organ and inter-organ dysregulation to be deconvoluted. When compared to normal fasting (NF) conditions, under early metabolic syndrome (EMS) conditions, the standalone vLAMPS exhibited characteristics of early stage MASLD, while no significant differences were observed in the standalone PANIS. In contrast, with EMS, the coupled vLAMPS-PANIS exhibited a perturbed islet-specific secretome and a significantly dysregulated glucose stimulated insulin secretion (GSIS) response implicating direct signaling from the dysregulated liver acinus to the islets. Correlations between several pairs of a vLAMPS-derived and a PANIS-derived secreted factors were significantly altered under EMS, as compared to NF conditions, mechanistically connecting MASLD and T2DM associated hepatic factors with islet-derived GLP-1 synthesis and regulation. Since vLAMPS-PANIS is compatible with patient-specific iPSCs, this platform represents an important step towards addressing patient heterogeneity, identifying complex disease mechanisms, and advancing precision medicine.
Collapse
|
19
|
Qin J, Qian Z, Lai Y, Zhang C, Zhang X. Microarray Platforms Based on 3D Printing. Anal Chem 2024; 96:6001-6011. [PMID: 38566481 DOI: 10.1021/acs.analchem.4c00367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
This paper introduces an innovative method for the fabrication and infusion of microwell arrays based on digital light processing (DLP) 3D printing. A low-cost DLP 3D printer is employed to fabricate microstructures rapidly with a broad dynamic range while maintaining high precision and fidelity. We constructed microwell arrays with varying diameters, from 200 to 2000 μm and multiple aspect ratios, in addition to microchannels with widths ranging from 45 to 1000 μm, proving the potential and flexibility of this fabrication method. The superimposition of parallel microchannels onto the microwell array, facilitated by positive or negative pressure, enabled the transfer of liquid to the microwells. Upon removal of the microchannel chip, a dispensed microdroplet array was obtained. This array can be modulated by adjusting the volume of the microwells and the inflow fluid. The filled microwell array allows chip-to-chip dispensing to the microreactor array through binding and centrifugation, facilitating multistep and multireagent assays. The 3D printing approach also enables the fabrication of intricate cavity designs, such as micropyramid arrays, which can be integrated with parallel microchannels to generate spheroid flowcells. This device demonstrated the ability to generate spheroids and manipulate their environment. We have successfully utilized precise modulation of spheroids size and performed parallel drug dose-response assays to evaluate its effectiveness. Furthermore, we managed to execute dynamic drug combinations based on a compact spheroids array, utilizing two orthogonal parallel microchannels. Our findings suggest that both the combination and temporal sequence of drug administration have a significant impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Jinglin Qin
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| | - Zhenwei Qian
- Peking University 302 Clinical Medical School, Beijing 100039, China
| | - Yiwen Lai
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Xiannian Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| |
Collapse
|
20
|
Schlünder K, Cipriano M, Zbinden A, Fuchs S, Mayr T, Schenke-Layland K, Loskill P. Microphysiological pancreas-on-chip platform with integrated sensors to model endocrine function and metabolism. LAB ON A CHIP 2024; 24:2080-2093. [PMID: 38441218 DOI: 10.1039/d3lc00838j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Pancreatic in vitro research is of major importance to advance mechanistic understanding and development of treatment options for diseases such as diabetes mellitus. We present a thermoplastic-based microphysiological system aiming to model the complex microphysiological structure and function of the endocrine pancreas with concurrent real-time read-out capabilities. The specifically tailored platform enables self-guided trapping of single islets at defined locations: β-cells are assembled to pseudo-islets and injected into the tissue chamber using hydrostatic pressure-driven flow. The pseudo-islets can further be embedded in an ECM-like hydrogel mimicking the native microenvironment of pancreatic islets in vivo. Non-invasive real-time monitoring of the oxygen levels on-chip is realized by the integration of luminescence-based optical sensors to the platform. To monitor insulin secretion kinetics in response to glucose stimulation in a time-resolved manner, an automated cycling of different glucose conditions is implemented. The model's response to glucose stimulation can be monitored via offline analysis of insulin secretion and via specific changes in oxygen consumption due to higher metabolic activity of pseudo-islets at high glucose levels. To demonstrate applicability for drug testing, the effects of antidiabetic medications are assessed and changes in dynamic insulin secretion are observed in line with the respective mechanism of action. Finally, by integrating human pancreatic islet microtissues, we highlight the flexibility of the platform and demonstrate the preservation of long-term functionality of human endocrine pancreatic tissue.
Collapse
Affiliation(s)
- Katharina Schlünder
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany.
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Aline Zbinden
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Stefanie Fuchs
- Institute for Analytical Chemistry and Food Chemistry, Graz University of Technology, Graz, Austria
| | - Torsten Mayr
- Institute for Analytical Chemistry and Food Chemistry, Graz University of Technology, Graz, Austria
| | - Katja Schenke-Layland
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany.
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Yang J, Yan Y, Yin X, Liu X, Reshetov IV, Karalkin PA, Li Q, Huang RL. Bioengineering and vascularization strategies for islet organoids: advancing toward diabetes therapy. Metabolism 2024; 152:155786. [PMID: 38211697 DOI: 10.1016/j.metabol.2024.155786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Diabetes presents a pressing healthcare crisis, necessitating innovative solutions. Organoid technologies have rapidly advanced, leading to the emergence of bioengineering islet organoids as an unlimited source of insulin-producing cells for treating insulin-dependent diabetes. This advancement surpasses the need for cadaveric islet transplantation. However, clinical translation of this approach faces two major limitations: immature endocrine function and the absence of a perfusable vasculature compared to primary human islets. In this review, we summarize the latest developments in bioengineering functional islet organoids in vitro and promoting vascularization of organoid grafts before and after transplantation. We highlight the crucial roles of the vasculature in ensuring long-term survival, maturation, and functionality of islet organoids. Additionally, we discuss key considerations that must be addressed before clinical translation of islet organoid-based therapy, including functional immaturity, undesired heterogeneity, and potential tumorigenic risks.
Collapse
Affiliation(s)
- Jing Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China
| | - Yuxin Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China
| | - Xiya Yin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China; Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, China
| | - Xiangqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China
| | - Igor V Reshetov
- Institute of Cluster Oncology, Sechenov First Moscow State Medical University, 127473 Moscow, Russia
| | - Pavel A Karalkin
- Institute of Cluster Oncology, Sechenov First Moscow State Medical University, 127473 Moscow, Russia
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China.
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China.
| |
Collapse
|
22
|
Nashimoto Y, Konno A, Imaizumi T, Nishikawa K, Ino K, Hori T, Kaji H, Shintaku H, Goto M, Shiku H. Microfluidic vascular formation model for assessing angiogenic capacities of single islets. Biotechnol Bioeng 2024; 121:1050-1059. [PMID: 38131167 DOI: 10.1002/bit.28631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/12/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Pancreatic islet transplantation presents a promising therapy for individuals suffering from type 1 diabetes. To maintain the function of transplanted islets in vivo, it is imperative to induce angiogenesis. However, the mechanisms underlying angiogenesis triggered by islets remain unclear. In this study, we introduced a microphysiological system to study the angiogenic capacity and dynamics of individual islets. The system, which features an open-top structure, uniquely facilitates the inoculation of islets and the longitudinal observation of vascular formation in in vivo like microenvironment with islet-endothelial cell communication. By leveraging our system, we discovered notable islet-islet heterogeneity in the angiogenic capacity. Transcriptomic analysis of the vascularized islets revealed that islets with high angiogenic capacity exhibited upregulation of genes related to insulin secretion and downregulation of genes related to angiogenesis and fibroblasts. In conclusion, our microfluidic approach is effective in characterizing the vascular formation of individual islets and holds great promise for elucidating the angiogenic mechanisms that enhance islet transplantation therapy.
Collapse
Affiliation(s)
- Yuji Nashimoto
- Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, Japan
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
- Cluster for Pioneering Research, RIKEN, Saitama, Japan
| | - An Konno
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
| | - Takuto Imaizumi
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
| | | | - Kosuke Ino
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
| | - Takeshi Hori
- Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokazu Kaji
- Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirofumi Shintaku
- Cluster for Pioneering Research, RIKEN, Saitama, Japan
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Hitoshi Shiku
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
| |
Collapse
|
23
|
Regeenes R, Rocheleau JV. Twenty years of islet-on-a-chip: microfluidic tools for dissecting islet metabolism and function. LAB ON A CHIP 2024; 24:1327-1350. [PMID: 38277011 DOI: 10.1039/d3lc00696d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Pancreatic islets are metabolically active micron-sized tissues responsible for controlling blood glucose through the secretion of insulin and glucagon. A loss of functional islet mass results in type 1 and 2 diabetes. Islet-on-a-chip devices are powerful microfluidic tools used to trap and study living ex vivo human and murine pancreatic islets and potentially stem cell-derived islet organoids. Devices developed over the past twenty years offer the ability to treat islets with controlled and dynamic microenvironments to mimic in vivo conditions and facilitate diabetes research. In this review, we explore the various islet-on-a-chip devices used to immobilize islets, regulate the microenvironment, and dynamically detect islet metabolism and insulin secretion. We first describe and assess the various methods used to immobilize islets including chambers, dam-walls, and hydrodynamic traps. We subsequently describe the surrounding methods used to create glucose gradients, enhance the reaggregation of dispersed islets, and control the microenvironment of stem cell-derived islet organoids. We focus on the various methods used to measure insulin secretion including capillary electrophoresis, droplet microfluidics, off-chip ELISAs, and on-chip fluorescence anisotropy immunoassays. Additionally, we delve into the various multiparametric readouts (NAD(P)H, Ca2+-activity, and O2-consumption rate) achieved primarily by adopting a microscopy-compatible optical window into the devices. By critical assessment of these advancements, we aim to inspire the development of new devices by the microfluidics community and accelerate the adoption of islet-on-a-chip devices by the wider diabetes research and clinical communities.
Collapse
Affiliation(s)
- Romario Regeenes
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Jonathan V Rocheleau
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Departments of Medicine and Physiology, University of Toronto, ON, Canada
| |
Collapse
|
24
|
Omori K, Qi M, Salgado M, Gonzalez N, Hui LT, Chen KT, Rawson J, Miao L, Komatsu H, Isenberg JS, Al-Abdullah IH, Mullen Y, Kandeel F. A scalable human islet 3D-culture platform maintains cell mass and function long-term for transplantation. Am J Transplant 2024; 24:177-189. [PMID: 37813189 DOI: 10.1016/j.ajt.2023.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023]
Abstract
Present-day islet culture methods provide short-term maintenance of cell viability and function, limiting access to islet transplantation. Attempts to lengthen culture intervals remain unsuccessful. A new method was developed to permit the long-term culture of islets. Human islets were embedded in polysaccharide 3D-hydrogel in cell culture inserts or gas-permeable chambers with serum-free CMRL 1066 supplemented media for up to 8 weeks. The long-term cultured islets maintained better morphology, cell mass, and viability at 4 weeks than islets in conventional suspension culture. In fact, islets cultured in the 3D-hydrogel retained β cell mass and function on par with freshly isolated islets in vitro and, when transplanted into diabetic mice, restored glucose balance similar to fresh islets. Using gas-permeable chambers, the 3D-hydrogel culture method was scaled up over 10-fold and maintained islet viability and function, although the cell mass recovery rate was 50%. Additional optimization of scale-up methods continues. If successful, this technology could afford flexibility and expand access to islet transplantation, especially single-donor islet-after-kidney transplantation.
Collapse
Affiliation(s)
- Keiko Omori
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Meirigeng Qi
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mayra Salgado
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Nelson Gonzalez
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Lauren T Hui
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Kuan-Tsen Chen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Lynn Miao
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hirotake Komatsu
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jeffrey S Isenberg
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yoko Mullen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
25
|
Shiwarski DJ, Hudson AR, Tashman JW, Bakirci E, Moss S, Coffin BD, Feinberg AW. 3D Bioprinting of Collagen-based Microfluidics for Engineering Fully-biologic Tissue Systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577422. [PMID: 38352326 PMCID: PMC10862740 DOI: 10.1101/2024.01.26.577422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Microfluidic and organ-on-a-chip devices have improved the physiologic and translational relevance of in vitro systems in applications ranging from disease modeling to drug discovery and pharmacology. However, current manufacturing approaches have limitations in terms of materials used, non-native mechanical properties, patterning of extracellular matrix (ECM) and cells in 3D, and remodeling by cells into more complex tissues. We present a method to 3D bioprint ECM and cells into microfluidic collagen-based high-resolution internally perfusable scaffolds (CHIPS) that address these limitations, expand design complexity, and simplify fabrication. Additionally, CHIPS enable size-dependent diffusion of molecules out of perfusable channels into the surrounding device to support cell migration and remodeling, formation of capillary-like networks, and integration of secretory cell types to form a glucose-responsive, insulin-secreting pancreatic-like microphysiological system.
Collapse
Affiliation(s)
- Daniel J. Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Andrew R. Hudson
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Joshua W. Tashman
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Samuel Moss
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Brian D. Coffin
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Adam W. Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Bender RHF, O’Donnell BT, Shergill B, Pham BQ, Tahmouresie S, Sanchez CN, Juat DJ, Hatch MMS, Shirure VS, Wortham M, Nguyen-Ngoc KV, Jun Y, Gaetani R, Christman KL, Teyton L, George SC, Sander M, Hughes CCW. A vascularized 3D model of the human pancreatic islet for ex vivostudy of immune cell-islet interaction. Biofabrication 2024; 16:025001. [PMID: 38128127 PMCID: PMC10782895 DOI: 10.1088/1758-5090/ad17d0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/24/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023]
Abstract
Insulin is an essential regulator of blood glucose homeostasis that is produced exclusively byβcells within the pancreatic islets of healthy individuals. In those affected by diabetes, immune inflammation, damage, and destruction of isletβcells leads to insulin deficiency and hyperglycemia. Current efforts to understand the mechanisms underlyingβcell damage in diabetes rely onin vitro-cultured cadaveric islets. However, isolation of these islets involves removal of crucial matrix and vasculature that supports islets in the intact pancreas. Unsurprisingly, these islets demonstrate reduced functionality over time in standard culture conditions, thereby limiting their value for understanding native islet biology. Leveraging a novel, vascularized micro-organ (VMO) approach, we have recapitulated elements of the native pancreas by incorporating isolated human islets within a three-dimensional matrix nourished by living, perfusable blood vessels. Importantly, these islets show long-term viability and maintain robust glucose-stimulated insulin responses. Furthermore, vessel-mediated delivery of immune cells to these tissues provides a model to assess islet-immune cell interactions and subsequent islet killing-key steps in type 1 diabetes pathogenesis. Together, these results establish the islet-VMO as a novel,ex vivoplatform for studying human islet biology in both health and disease.
Collapse
Affiliation(s)
- R Hugh F Bender
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Benjamen T O’Donnell
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Bhupinder Shergill
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Brittany Q Pham
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Sima Tahmouresie
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Celeste N Sanchez
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Damie J Juat
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Michaela M S Hatch
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Matthew Wortham
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, CA, United States of America
| | - Kim-Vy Nguyen-Ngoc
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, CA, United States of America
| | - Yesl Jun
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, CA, United States of America
| | - Roberto Gaetani
- Department of Bioengineering, University of California, San Diego, CA, United States of America
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Karen L Christman
- Department of Cellular & Molecular Medicine, University of California, San Diego, CA, United States of America
- Department of Bioengineering, University of California, San Diego, CA, United States of America
| | - Luc Teyton
- Department of Immunology & Microbiology, The Scripps Research Institute, San Diego, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Maike Sander
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, CA, United States of America
- Department of Cellular & Molecular Medicine, University of California, San Diego, CA, United States of America
| | - Christopher C W Hughes
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California, Irvine, CA, United States of America
| |
Collapse
|
27
|
Oppler SH, Hocum Stone LL, Leishman DJ, Janecek JL, Moore MEG, Rangarajan P, Willenberg BJ, O’Brien TD, Modiano J, Pheil N, Dalton J, Dalton M, Ramachandran S, Graham ML. A bioengineered artificial interstitium supports long-term islet xenograft survival in nonhuman primates without immunosuppression. SCIENCE ADVANCES 2024; 10:eadi4919. [PMID: 38181083 PMCID: PMC10776017 DOI: 10.1126/sciadv.adi4919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 12/02/2023] [Indexed: 01/07/2024]
Abstract
Cell-based therapies hold promise for many chronic conditions; however, the continued need for immunosuppression along with challenges in replacing cells to improve durability or retrieving cells for safety are major obstacles. We subcutaneously implanted a device engineered to exploit the innate transcapillary hydrostatic and colloid osmotic pressure generating ultrafiltrate to mimic interstitium. Long-term stable accumulation of ultrafiltrate was achieved in both rodents and nonhuman primates (NHPs) that was chemically similar to serum and achieved capillary blood oxygen concentration. The majority of adult pig islet grafts transplanted in non-immunosuppressed NHPs resulted in xenograft survival >100 days. Stable cytokine levels, normal neutrophil to lymphocyte ratio, and a lack of immune cell infiltration demonstrated successful immunoprotection and averted typical systemic changes related to xenograft transplant, especially inflammation. This approach eliminates the need for immunosuppression and permits percutaneous access for loading, reloading, biopsy, and recovery to de-risk the use of "unlimited" xenogeneic cell sources to realize widespread clinical translation of cell-based therapies.
Collapse
Affiliation(s)
- Scott H. Oppler
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | | | - David J. Leishman
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Jody L. Janecek
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Meghan E. G. Moore
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
| | | | - Bradley J. Willenberg
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Timothy D. O’Brien
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Jaime Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
| | - Natan Pheil
- Cell-Safe LifeSciences, Skokie, IL, USA
- Medline UNITE Foot and Ankle, Medline Industries LP, 3 Lakes Drive, Northfield, IL, USA
| | | | | | | | - Melanie L. Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| |
Collapse
|
28
|
Wei J, Yang Y, Li M. Single-cell force spectroscopy of fluid flow-tuned cell adhesion for dissecting hemodynamics in tumor metastasis. NANOSCALE 2023; 16:360-372. [PMID: 38063483 DOI: 10.1039/d3nr04439d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Cell adhesion plays an important role in regulating the metastasis of cancer cells, and atomic force microscopy (AFM)-based single-cell force spectroscopy (SCFS) has become an important method to directly measure the adhesion forces of individual cells. Particularly, bodily fluid flow environments strongly affect the functions and behaviors of metastatic cells for successful dissemination. Nevertheless, the interactions between fluidic flow medium environment and cell adhesion remain poorly understood. In this work, AFM-based SCFS was exploited to examine the effects of fluidic flow environment on cellular adhesion. A fluidic cell culture medium device was used to simulate the fluidic flow environment experienced by cancer cells during metastasis, which was combined with AFM-based SCFS assay. A single living cancer cell was attached to the AFM tipless cantilever to prepare the single-cell probe for performing SCFS experiments on the mesothelial cells grown under the fluidic flow medium conditions, and the effects of experimental parameters (retraction speed, contact time, loading force) on the measured cellular adhesion forces were analyzed. Experimental results of SCFS assay show that cellular adhesion forces significantly decrease after growth in fluidic flow medium, whereas cellular adhesion forces increase after growth in static culture medium. Experiments performed with the use of spherical probes coated with cell adhesion-associated biomolecules also show the weakening of cell adhesion after growth in fluidic flow cell culture medium, which was subsequently confirmed by the confocal fluorescence microscopy experiments of cell adhesion molecules, vividly illustrating the remarkable effects of fluidic flow environment on cellular adhesion. The study provides a new approach to detect adhesion force dynamics involved in the interactions between cells and the fluidic flow environment at the single-cell level, which will facilitate dissecting the role of hemodynamics in tumor metastasis.
Collapse
Affiliation(s)
- Jiajia Wei
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanqi Yang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
29
|
Tronolone JJ, Mathur T, Chaftari CP, Sun Y, Jain A. Machine learning chained neural network analysis of oxygen transport amplifies the physiological relevance of vascularized microphysiological systems. Bioeng Transl Med 2023; 8:e10582. [PMID: 38023704 PMCID: PMC10658488 DOI: 10.1002/btm2.10582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/04/2023] [Accepted: 07/14/2023] [Indexed: 12/01/2023] Open
Abstract
Since every biological system requires capillaries to support its oxygenation, design of engineered preclinical models of such systems, for example, vascularized microphysiological systems (vMPS) have gained attention enhancing the physiological relevance of human biology and therapies. But the physiology and function of formed vessels in the vMPS is currently assessed by non-standardized, user-dependent, and simple morphological metrics that poorly relate to the fundamental function of oxygenation of organs. Here, a chained neural network is engineered and trained using morphological metrics derived from a diverse set of vMPS representing random combinations of factors that influence the vascular network architecture of a tissue. This machine-learned algorithm outputs a singular measure, termed as vascular network quality index (VNQI). Cross-correlation of morphological metrics and VNQI against measured oxygen levels within vMPS revealed that VNQI correlated the most with oxygen measurements. VNQI is sensitive to the determinants of vascular networks and it consistently correlates better to the measured oxygen than morphological metrics alone. Finally, the VNQI is positively associated with the functional outcomes of cell transplantation therapies, shown in the vascularized islet-chip challenged with hypoxia. Therefore, adoption of this tool will amplify the predictions and enable standardization of organ-chips, transplant models, and other cell biosystems.
Collapse
Affiliation(s)
- James J. Tronolone
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Christopher P. Chaftari
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
- Department of Medical Physiology, School of MedicineTexas A&M Health Science CenterBryanTexasUSA
- Department of Cardiovascular ScienceHouston Methodist Academic InstituteHoustonTexasUSA
| |
Collapse
|
30
|
Vanderlaan EL, Sexton J, Evans-Molina C, Buganza Tepole A, Voytik-Harbin SL. Islet-on-chip: promotion of islet health and function via encapsulation within a polymerizable fibrillar collagen scaffold. LAB ON A CHIP 2023; 23:4466-4482. [PMID: 37740372 DOI: 10.1039/d3lc00371j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
The protection and interrogation of pancreatic β-cell health and function ex vivo is a fundamental aspect of diabetes research, including mechanistic studies, evaluation of β-cell health modulators, and development and quality control of replacement β-cell populations. However, present-day islet culture formats, including traditional suspension culture as well as many recently developed microfluidic devices, suspend islets in a liquid microenvironment, disrupting mechanochemical signaling normally found in vivo and limiting β-cell viability and function in vitro. Herein, we present a novel three-dimensional (3D) microphysiological system (MPS) to extend islet health and function ex vivo by incorporating a polymerizable collagen scaffold to restore biophysical support and islet-collagen mechanobiological cues. Informed by computational models of gas and molecular transport relevant to β-cell physiology, a MPS configuration was down-selected based on simulated oxygen and nutrient delivery to collagen-encapsulated islets, and 3D-printing was applied as a readily accessible, low-cost rapid prototyping method. Recreating critical aspects of the in vivo microenvironment within the MPS via perfusion and islet-collagen interactions mitigated post-isolation ischemia and apoptosis in mouse islets over a 5-day period. In contrast, islets maintained in traditional suspension formats exhibited progressive hypoxic and apoptotic cores. Finally, dynamic glucose-stimulated insulin secretion measurements were performed on collagen-encapsulated mouse islets in the absence and presence of well-known chemical stressor thapsigargin using the MPS platform and compared to conventional protocols involving commercial perifusion machines. Overall, the MPS described here provides a user-friendly islet culture platform that not only supports long-term β-cell health and function but also enables multiparametric evaluations.
Collapse
Affiliation(s)
- Emma L Vanderlaan
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Medical Scientist/Engineer Training Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joshua Sexton
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Adrian Buganza Tepole
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA.
- School of Mechanical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| |
Collapse
|
31
|
Sun S, Xue X, Fu J. Modeling development using microfluidics: bridging gaps to foster fundamental and translational research. Curr Opin Genet Dev 2023; 82:102097. [PMID: 37573835 PMCID: PMC11193336 DOI: 10.1016/j.gde.2023.102097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/01/2023] [Accepted: 07/16/2023] [Indexed: 08/15/2023]
Abstract
In vitro stem cell-derived embryo and organ models, termed embryoids and organoids, respectively, provide promising experimental tools to study physiological and pathological processes in mammalian development and organ formation. Most of current embryoid and organoid systems are developed using conventional three-dimensional cultures that lack controls of spatiotemporal extracellular signals. Microfluidics, an established technology for quantitative controls and quantifications of dynamic chemical and physical environments, has recently been utilized for developing next-generation embryoids and organoids in a controllable and reproducible manner. In this review, we summarize recent progress in constructing microfluidics-based embryoids and organoids. Development of these models demonstrates the successful applications of microfluidics in establishing morphogen gradients, accelerating medium transport, exerting mechanical forces, facilitating tissue coculture studies, and improving assay throughput, thus supporting using microfluidics for building next-generation embryoids and organoids for fundamental and translational research.
Collapse
Affiliation(s)
- Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
Xu Y, Zhou A, Chen W, Ning X. Scaffold-Free Multicellular 3D Tissue Constructs Utilizing Bio-orthogonal Click Strategy. NANO LETTERS 2023; 23:8770-8778. [PMID: 37694972 DOI: 10.1021/acs.nanolett.3c02889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Multicellular 3D tissue constructs (MTCs) are important in biomedical research due to their capacity to accurately mimic the structure and variation found in real tissues. This study presents a novel bio-orthogonal engineering strategy (BIEN), a transformative scaffold-free approach, to create advanced MTCs. BIEN harnesses the cellular biosynthetic machinery to incorporate bio-orthogonal azide reporters into cell surface glycoconjugates, followed by a click reaction with multiarm PEG, resulting in rapid assembly of MTCs. The implementation of this cutting-edge strategy culminates in the formation of uniform, heterogeneous spheroids, characterized by a high degree of intercellular junction and pluripotency. Remarkably, MTCs simulate tumor features, ensure cell heterogeneity, and significantly improve the subcutaneous xenograft model after transplantation, thereby bolstering both in vitro and in vivo research models. In conclusion, the utilization of the bio-orthogonal engineering strategy as a scaffold-free method to generate superior MTCs holds promising potential for driving advancements in cancer research.
Collapse
Affiliation(s)
- Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing 210093, People's Republic of China
| | - Weiwei Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| |
Collapse
|
33
|
Vanderlaan EL, Nolan JK, Sexton J, Evans-Molina C, Lee H, Voytik-Harbin SL. Development of electrochemical Zn 2+ sensors for rapid voltammetric detection of glucose-stimulated insulin release from pancreatic β-cells. Biosens Bioelectron 2023; 235:115409. [PMID: 37244091 DOI: 10.1016/j.bios.2023.115409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023]
Abstract
Diabetes is a chronic disease characterized by elevated blood glucose levels resulting from absent or ineffective insulin release from pancreatic β-cells. β-cell function is routinely assessed in vitro using static or dynamic glucose-stimulated insulin secretion (GSIS) assays followed by insulin quantification via time-consuming, costly enzyme-linked immunosorbent assays (ELISA). In this study, we developed a highly sensitive electrochemical sensor for zinc (Zn2+), an ion co-released with insulin, as a rapid and low-cost method for measuring dynamic insulin release. Different modifications to glassy carbon electrodes (GCE) were evaluated to develop a sensor that detects physiological Zn2+ concentrations while operating within a biological Krebs Ringer Buffer (KRB) medium (pH 7.2). Electrodeposition of bismuth and indium improved Zn2+ sensitivity and limit of detection (LOD), and a Nafion coating improved selectivity. Using anodic stripping voltammetry (ASV) with a pre-concentration time of 6 min, we achieved a LOD of 2.3 μg/L over the wide linear range of 2.5-500 μg/L Zn2+. Sensor performance improved with 10-min pre-concentration, resulting in increased sensitivity, lower LOD (0.18 μg/L), and a bilinear response over the range of 0.25-10 μg/L Zn2+. We further characterized the physicochemical properties of the Zn2+ sensor using scanning electron microscopy (SEM), cyclic voltammetry (CV), and electrochemical impedance spectroscopy (EIS). Finally, we demonstrated the sensor's capability to measure Zn2+ release from glucose-stimulated INS-1 β-cells and primary mouse islets. Our results exhibited a high correlation with secreted insulin and validated the sensor's potential as a rapid alternative to conventional two-step GSIS plus ELISA methods.
Collapse
Affiliation(s)
- Emma L Vanderlaan
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA; Indiana Medical Scientist/Engineer Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James K Nolan
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA; Center for Implantable Devices, Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
| | - Joshua Sexton
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA
| | - Carmella Evans-Molina
- Indiana Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Hyowon Lee
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA; Center for Implantable Devices, Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA; Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
34
|
Li H, Shang Y, Feng Q, Liu Y, Chen J, Dong H. A novel bioartificial pancreas fabricated via islets microencapsulation in anti-adhesive core-shell microgels and macroencapsulation in a hydrogel scaffold prevascularized in vivo. Bioact Mater 2023; 27:362-376. [PMID: 37180642 PMCID: PMC10172916 DOI: 10.1016/j.bioactmat.2023.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Islets transplantation is a promising treatment for type 1 diabetes mellitus. However, severe host immune rejection and poor oxygen/nutrients supply due to the lack of surrounding capillary network often lead to transplantation failure. Herein, a novel bioartificial pancreas is constructed via islets microencapsulation in core-shell microgels and macroencapsulation in a hydrogel scaffold prevascularized in vivo. Specifically, a hydrogel scaffold containing methacrylated gelatin (GelMA), methacrylated heparin (HepMA) and vascular endothelial growth factor (VEGF) is fabricated, which can delivery VEGF in a sustained style and thus induce subcutaneous angiogenesis. In addition, islets-laden core-shell microgels using methacrylated hyaluronic acid (HAMA) as microgel core and poly(ethylene glycol) diacrylate (PEGDA)/carboxybetaine methacrylate (CBMA) as shell layer are prepared, which provide a favorable microenvironment for islets and simultaneously the inhibition of host immune rejection via anti-adhesion of proteins and immunocytes. As a result of the synergistic effect between anti-adhesive core-shell microgels and prevascularized hydrogel scaffold, the bioartificial pancreas can reverse the blood glucose levels of diabetic mice from hyperglycemia to normoglycemia for at least 90 days. We believe this bioartificial pancreas and relevant fabrication method provide a new strategy to treat type 1 diabetes, and also has broad potential applications in other cell therapies.
Collapse
Affiliation(s)
- Haofei Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Yulian Shang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Qi Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Yang Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Junlin Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Hua Dong
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
35
|
Takagi M, Yamada M, Utoh R, Seki M. A multiscale, vertical-flow perfusion system with integrated porous microchambers for upgrading multicellular spheroid culture. LAB ON A CHIP 2023; 23:2257-2267. [PMID: 37038847 DOI: 10.1039/d3lc00168g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Spheroid formation assisted by microengineered chambers is a versatile approach for morphology-controlled three-dimensional (3D) cell cultivation with physiological relevance to human tissues. However, the limitation in diffusion-based oxygen/nutrient transport has been a critical issue for the densely packed cells in spheroids, preventing maximization of cellular functions and thus limiting their biomedical applications. Here, we have developed a multiscale microfluidic system for the perfusion culture of spheroids, in which porous microchambers, connected with microfluidic channels, were engineered. A newly developed process of centrifugation-assisted replica molding and salt-leaching enabled the formation of single micrometer-sized pores on the chamber surface and in the substrate. The porous configuration generates a vertical flow to directly supply the medium to the spheroids, while avoiding the formation of stagnant flow regions. We created seamlessly integrated, all PDMS/silicone-based microfluidic devices with an array of microchambers. Spheroids of human liver cells (HepG2 cells) were formed and cultured under vertical-flow perfusion, and the proliferation ability and liver cell-specific functions were compared with those of cells cultured in non-porous chambers with a horizontal flow. The presented system realizes both size-controlled formation of spheroids and direct medium supply, making it suitable as a precision cell culture platform for drug development, disease modelling, and regenerative medicine.
Collapse
Affiliation(s)
- Mai Takagi
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| |
Collapse
|
36
|
Do TD, Pham UT, Nguyen LP, Nguyen TM, Bui CN, Oliver S, Pham P, Tran TQ, Hoang BT, Pham MTH, Pham DTN, Nguyen DT. Fabrication of a Low-Cost Microfluidic Device for High-Throughput Drug Testing on Static and Dynamic Cancer Spheroid Culture Models. Diagnostics (Basel) 2023; 13:diagnostics13081394. [PMID: 37189495 DOI: 10.3390/diagnostics13081394] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/17/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Drug development is a complex and expensive process from new drug discovery to product approval. Most drug screening and testing rely on in vitro 2D cell culture models; however, they generally lack in vivo tissue microarchitecture and physiological functionality. Therefore, many researchers have used engineering methods, such as microfluidic devices, to culture 3D cells in dynamic conditions. In this study, a simple and low-cost microfluidic device was fabricated using Poly Methyl Methacrylate (PMMA), a widely available material, and the total cost of the completed device was USD 17.75. Dynamic and static cell culture examinations were applied to monitor the growth of 3D cells. α-MG-loaded GA liposomes were used as the drug to test cell viability in 3D cancer spheroids. Two cell culture conditions (i.e., static and dynamic) were also used in drug testing to simulate the effect of flow on drug cytotoxicity. Results from all assays showed that with the velocity of 0.005 mL/min, cell viability was significantly impaired to nearly 30% after 72 h in a dynamic culture. This device is expected to improve in vitro testing models, reduce and eliminate unsuitable compounds, and select more accurate combinations for in vivo testing.
Collapse
Affiliation(s)
- Tung Dinh Do
- Saint Paul General Hospital, No. 12, Chu Van An St., Ba Dinh Dist, Ha Noi 10000, Vietnam
| | - Uyen Thu Pham
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Linh Phuong Nguyen
- School of Preventive Medicine and Public Health, Hanoi Medical University, 1 Ton That Tung St., Dong Da Dist., Hanoi 10000, Vietnam
| | - Trang Minh Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Cuong Nguyen Bui
- Hung Yen University of Technology and Education (UTEHY), 39A St., Khoai Chau Dist., Hung Yen 17000, Vietnam
| | - Susan Oliver
- Centre for Advanced Macromolecular Design and Australian Centre for NanoMedicine, School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Phuong Pham
- Centre for Advanced Macromolecular Design and Australian Centre for NanoMedicine, School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Toan Quoc Tran
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Bich Thi Hoang
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Minh Thi Hong Pham
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Duong Thanh Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| |
Collapse
|
37
|
Beydag-Tasöz BS, Yennek S, Grapin-Botton A. Towards a better understanding of diabetes mellitus using organoid models. Nat Rev Endocrinol 2023; 19:232-248. [PMID: 36670309 PMCID: PMC9857923 DOI: 10.1038/s41574-022-00797-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/22/2023]
Abstract
Our understanding of diabetes mellitus has benefited from a combination of clinical investigations and work in model organisms and cell lines. Organoid models for a wide range of tissues are emerging as an additional tool enabling the study of diabetes mellitus. The applications for organoid models include studying human pancreatic cell development, pancreatic physiology, the response of target organs to pancreatic hormones and how glucose toxicity can affect tissues such as the blood vessels, retina, kidney and nerves. Organoids can be derived from human tissue cells or pluripotent stem cells and enable the production of human cell assemblies mimicking human organs. Many organ mimics relevant to diabetes mellitus are already available, but only a few relevant studies have been performed. We discuss the models that have been developed for the pancreas, liver, kidney, nerves and vasculature, how they complement other models, and their limitations. In addition, as diabetes mellitus is a multi-organ disease, we highlight how a merger between the organoid and bioengineering fields will provide integrative models.
Collapse
Affiliation(s)
- Belin Selcen Beydag-Tasöz
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Siham Yennek
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
| | - Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Paul Langerhans Institute Dresden, Dresden, Germany.
| |
Collapse
|
38
|
Kim SY, Ha SM, Kim DU, Park J, Park S, Hyun KA, Jung HI. Modularized dynamic cell culture platform for efficient production of extracellular vesicles and sequential analysis. LAB ON A CHIP 2023; 23:1852-1864. [PMID: 36825402 DOI: 10.1039/d2lc01129h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Extracellular vesicles (EVs) are nanometer-sized particles naturally secreted by cells for intercellular communication that encapsulate bioactive cargo, such as proteins and RNA, with a lipid bilayer. Tumor cell-derived EVs (tdEVs) are particularly promising biomarkers for cancer research because their contents reflect the cell of origin. In most studies, tdEVs have been obtained from cancer cells cultured under static conditions, thus lacking the ability to recapitulate the microenvironment of cells in vivo. Recent developments in perfusable cell culture systems have allowed oxygen and a nutrient gradient to mimic the physiological and cellular microenvironment. However, as these systems are perfused by circulating the culture medium within the unified structure, independently harvesting cells and EVs at each time point for analysis presents a limitation. In this study, a modularized cell culture system is designed for the perfusion and real-time collection of EVs. The system consists of three detachable chambers, one each for fresh medium, cell culture, and EV collection. The fresh medium flows from the medium chamber to the culture chamber at a flow rate controlled by the hydraulic pressure injected with a syringe pump. When the culture medium containing EVs exceeds a certain volume within the chamber, it overflows into the collection chamber to harvest EVs. The compact and modularized chambers are highly interoperable with conventional cell culture modalities used in the laboratory, thus enabling various EV-based assays.
Collapse
Affiliation(s)
- Seo Yeon Kim
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
| | - Seong Min Ha
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
| | - Dong-Uk Kim
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
| | - Junhyun Park
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
| | - Sunyoung Park
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
- The DABOM Inc., 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea
| | - Kyung-A Hyun
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
- The DABOM Inc., 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea
| |
Collapse
|
39
|
From Single- to Multi-organ-on-a-Chip System for Studying Metabolic Diseases. BIOCHIP JOURNAL 2023. [DOI: 10.1007/s13206-023-00098-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
40
|
Goldrick C, Guri I, Herrera-Oropeza G, O’Brien-Gore C, Roy E, Wojtynska M, Spagnoli FM. 3D multicellular systems in disease modelling: From organoids to organ-on-chip. Front Cell Dev Biol 2023; 11:1083175. [PMID: 36819106 PMCID: PMC9933985 DOI: 10.3389/fcell.2023.1083175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Cell-cell interactions underlay organ formation and function during homeostasis. Changes in communication between cells and their surrounding microenvironment are a feature of numerous human diseases, including metabolic disease and neurological disorders. In the past decade, cross-disciplinary research has been conducted to engineer novel synthetic multicellular organ systems in 3D, including organoids, assembloids, and organ-on-chip models. These model systems, composed of distinct cell types, satisfy the need for a better understanding of complex biological interactions and mechanisms underpinning diseases. In this review, we discuss the emerging field of building 3D multicellular systems and their application for modelling the cellular interactions at play in diseases. We report recent experimental and computational approaches for capturing cell-cell interactions as well as progress in bioengineering approaches for recapitulating these complexities ex vivo. Finally, we explore the value of developing such multicellular systems for modelling metabolic, intestinal, and neurological disorders as major examples of multisystemic diseases, we discuss the advantages and disadvantages of the different approaches and provide some recommendations for further advancing the field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francesca M. Spagnoli
- Faculty of Life Sciences, Centre for Gene Therapy and Regenerative Medicine, Guy’s Campus, King’s College London, London, United Kingdom
| |
Collapse
|
41
|
Li W, Peng YF. Advances in microfluidic chips based on islet hormone-sensing techniques. World J Diabetes 2023; 14:17-25. [PMID: 36684385 PMCID: PMC9850799 DOI: 10.4239/wjd.v14.i1.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/11/2022] [Accepted: 12/07/2022] [Indexed: 01/10/2023] Open
Abstract
Diabetes mellitus is a global health problem resulting from islet dysfunction or insulin resistance. The mechanisms of islet dysfunction are still under investigation. Islet hormone secretion is the main function of islets, and serves an important role in the homeostasis of blood glucose. Elucidating the detailed mechanism of islet hormone secretome distortion can provide clues for the treatment of diabetes. Therefore, it is crucial to develop accurate, real-time, labor-saving, high-throughput, automated, and cost-effective techniques for the sensing of islet secretome. Microfluidic chips, an elegant platform that combines biology, engineering, computer science, and biomaterials, have attracted tremendous interest from scientists in the field of diabetes worldwide. These tiny devices are miniatures of traditional experimental systems with more advantages of time-saving, reagent-minimization, automation, high-throughput, and online detection. These features of microfluidic chips meet the demands of islet secretome analysis and a variety of chips have been designed in the past 20 years. In this review, we present a brief introduction of microfluidic chips, and three microfluidic chips-based islet hormone sensing techniques. We focus mainly on the theory of these techniques, and provide detailed examples based on these theories with the hope of providing some insights into the design of future chips or whole detection systems.
Collapse
Affiliation(s)
- Wei Li
- Department of Endocrinology, Suzhou Hospital of Anhui Medical University, Suzhou 234000, Anhui Province, China
| | - You-Fan Peng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
42
|
Rodrigues Oliveira SM, Rebocho A, Ahmadpour E, Nissapatorn V, de Lourdes Pereira M. Type 1 Diabetes Mellitus: A Review on Advances and Challenges in Creating Insulin Producing Devices. MICROMACHINES 2023; 14:151. [PMID: 36677212 PMCID: PMC9867263 DOI: 10.3390/mi14010151] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/25/2022] [Accepted: 12/30/2022] [Indexed: 06/17/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is the most common autoimmune chronic disease in young patients. It is caused by the destruction of pancreatic endocrine β-cells that produce insulin in specific areas of the pancreas, known as islets of Langerhans. As a result, the body becomes insulin deficient and hyperglycemic. Complications associated with diabetes are life-threatening and the current standard of care for T1DM consists still of insulin injections. Lifesaving, exogenous insulin replacement is a chronic and costly burden of care for diabetic patients. Alternative therapeutic options have been the focus in these fields. Advances in molecular biology technologies and in microfabrication have enabled promising new therapeutic options. For example, islet transplantation has emerged as an effective treatment to restore the normal regulation of blood glucose in patients with T1DM. However, this technique has been hampered by obstacles, such as limited islet availability, extensive islet apoptosis, and poor islet vascular engraftment. Many of these unsolved issues need to be addressed before a potential cure for T1DM can be a possibility. New technologies like organ-on-a-chip platforms (OoC), multiplexed assessment tools and emergent stem cell approaches promise to enhance therapeutic outcomes. This review will introduce the disorder of type 1 diabetes mellitus, an overview of advances and challenges in the areas of microfluidic devices, monitoring tools, and prominent use of stem cells, and how they can be linked together to create a viable model for the T1DM treatment. Microfluidic devices like OoC platforms can establish a crucial platform for pathophysiological and pharmacological studies as they recreate the pancreatic environment. Stem cell use opens the possibility to hypothetically generate a limitless number of functional pancreatic cells. Additionally, the integration of stem cells into OoC models may allow personalized or patient-specific therapies.
Collapse
Affiliation(s)
- Sonia M. Rodrigues Oliveira
- HMRI-Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - António Rebocho
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ehsan Ahmadpour
- Drug Applied Research Center, Department of Parasitology and Mycology, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Veeranoot Nissapatorn
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Allied Health Sciences, Southeast Asia Water Team (SEAWater Team), World Union for Herbal Drug Discovery (WUHeDD), Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
43
|
Essaouiba A, Jellali R, Gilard F, Gakière B, Okitsu T, Legallais C, Sakai Y, Leclerc E. Investigation of the Exometabolomic Profiles of Rat Islets of Langerhans Cultured in Microfluidic Biochip. Metabolites 2022; 12:metabo12121270. [PMID: 36557308 PMCID: PMC9786643 DOI: 10.3390/metabo12121270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus (DM) is a complex disease with high prevalence of comorbidity and mortality. DM is predicted to reach more than 700 million people by 2045. In recent years, several advanced in vitro models and analytical tools were developed to investigate the pancreatic tissue response to pathological situations and identify therapeutic solutions. Of all the in vitro promising models, cell culture in microfluidic biochip allows the reproduction of in-vivo-like micro-environments. Here, we cultured rat islets of Langerhans using dynamic cultures in microfluidic biochips. The dynamic cultures were compared to static islets cultures in Petri. The islets' exometabolomic signatures, with and without GLP1 and isradipine treatments, were characterized by GC-MS. Compared to Petri, biochip culture contributes to maintaining high secretions of insulin, C-peptide and glucagon. The exometabolomic profiling revealed 22 and 18 metabolites differentially expressed between Petri and biochip on Day 3 and 5. These metabolites illustrated the increase in lipid metabolism, the perturbation of the pentose phosphate pathway and the TCA cycle in biochip. After drug stimulations, the exometabolome of biochip culture appeared more perturbed than the Petri exometabolome. The GLP1 contributed to the increase in the levels of glycolysis, pentose phosphate and glutathione pathways intermediates, whereas isradipine led to reduced levels of lipids and carbohydrates.
Collapse
Affiliation(s)
- Amal Essaouiba
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Rachid Jellali
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
- Correspondence: (R.J.); (E.L.)
| | - Françoise Gilard
- Plateforme Métabolisme-Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université Evry, Université Paris Cité, Bâtiment 360, Avenue des Sciences, 91190 Gif sur Yvette, France
| | - Bertrand Gakière
- Plateforme Métabolisme-Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université Evry, Université Paris Cité, Bâtiment 360, Avenue des Sciences, 91190 Gif sur Yvette, France
| | - Teru Okitsu
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Cécile Legallais
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
| | - Yasuyuki Sakai
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Eric Leclerc
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Correspondence: (R.J.); (E.L.)
| |
Collapse
|
44
|
Olaniru OE, Hook P, Persaud SJ. Using single-cell multi-omics screening of human fetal pancreas to identify novel players in human beta cell development. Diabet Med 2022; 39:e14992. [PMID: 36302085 PMCID: PMC9828353 DOI: 10.1111/dme.14992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/20/2022] [Indexed: 01/18/2023]
Abstract
Islet transplantation from organ donors can considerably improve glucose homeostasis and well-being in individuals with type 1 diabetes, where the beta cells are destroyed by the autoimmune attack, but there are insufficient donor islets to make this a widespread therapy. Strategies are therefore being developed to generate unlimited amounts of insulin-producing beta cells from pluripotent stem cells, with the aim that they will be transplanted to treat diabetes. Whilst much progress has been made in recent years in the directed differentiation of pluripotent stem cells to beta-like cells, essential gaps still exist in generating stem cell-derived beta cells that are fully functional in vitro. This short review provides details of recent multi-'omics' studies of the human fetal pancreas, which are revealing granular information on the various cell types in the developing pancreas. It is anticipated that this fine mapping of the pancreatic cells at single-cell resolution will provide additional insights that can be utilised to reproducibly produce human beta cells in vitro that have the functional characteristics of beta cells within native human islets.
Collapse
Affiliation(s)
- Oladapo E. Olaniru
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Philippa Hook
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Shanta J. Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| |
Collapse
|
45
|
Kato H, Miwa T, Quijano J, Medrano L, Ortiz J, Desantis A, Omori K, Wada A, Tatsukoshi K, Kandeel F, Mullen Y, Ku HT, Komatsu H. Microwell culture platform maintains viability and mass of human pancreatic islets. Front Endocrinol (Lausanne) 2022; 13:1015063. [PMID: 36465665 PMCID: PMC9712283 DOI: 10.3389/fendo.2022.1015063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Background Transplantation of the human pancreatic islets is a promising approach for specific types of diabetes to improve glycemic control. Although effective, there are several issues that limit the clinical expansion of this treatment, including difficulty in maintaining the quality and quantity of isolated human islets prior to transplantation. During the culture, we frequently observe the multiple islets fusing together into large constructs, in which hypoxia-induced cell damage significantly reduces their viability and mass. In this study, we introduce the microwell platform optimized for the human islets to prevent unsolicited fusion, thus maintaining their viability and mass in long-term cultures. Method Human islets are heterogeneous in size; therefore, two different-sized microwells were prepared in a 35 mm-dish format: 140 µm × 300 µm-microwells for <160 µm-islets and 200 µm × 370 µm-microwells for >160 µm-islets. Human islets (2,000 islet equivalent) were filtered through a 160 µm-mesh to prepare two size categories for subsequent two week-cultures in each microwell dish. Conventional flat-bottomed 35 mm-dishes were used for non-filtered islets (2,000 islet equivalent/2 dishes). Post-cultured islets are collected to combine in each condition (microwells and flat) for the comparisons in viability, islet mass, morphology, function and metabolism. Islets from three donors were independently tested. Results The microwell platform prevented islet fusion during culture compared to conventional flat bottom dishes, which improved human islet viability and mass. Islet viability and mass on the microwells were well-maintained and comparable to those in pre-culture, while flat bottom dishes significantly reduced islet viability and mass in two weeks. Morphology assessed by histology, insulin-secreting function and metabolism by oxygen consumption did not exhibit the statistical significance among the three different conditions. Conclusion Microwell-bottomed dishes maintained viability and mass of human islets for two weeks, which is significantly improved when compared to the conventional flat-bottomed dishes.
Collapse
Affiliation(s)
- Hiroyuki Kato
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | | | - Janine Quijano
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Leonard Medrano
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Jose Ortiz
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Akiko Desantis
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Keiko Omori
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Aya Wada
- AGC Techno Glass, Shizuoka, Japan
| | | | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Yoko Mullen
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Hsun Teresa Ku
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Hirotake Komatsu
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute of City of Hope, Duarte, CA, United States
| |
Collapse
|
46
|
Goswami I, de Klerk E, Carnese P, Hebrok M, Healy KE. Multiplexed microfluidic platform for stem-cell derived pancreatic islet β cells. LAB ON A CHIP 2022; 22:4430-4442. [PMID: 36305868 PMCID: PMC9642094 DOI: 10.1039/d2lc00468b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
Stem cell-derived β cells offer an alternative to primary islets for biomedical discoveries as well as a potential surrogate for islet transplantation. The expense and challenge of obtaining and maintaining functional stem cell-derived β cells calls for a need to develop better high-content and high-throughput culture systems. Microphysiological systems (MPS) are promising high-content in vitro platforms, but scaling for high-throughput screening and discoveries remain a challenge. Traditionally, simultaneous multiplexing of liquid handling and cell loading poses a challenge in the design of high-throughput MPS. Furthermore, although MPS for islet β culture/testing have been developed, studies on multi-day culture of stem-cell derived β cells in MPS have been limited. We present a scalable, multiplexed islet β MPS device that incorporates microfluidic gradient generators to parallelize fluid handling for culture and test conditions. We demonstrated the viability and functionality of the stem cell-derived enriched β clusters (eBCs) for a week, as assessed by the ∼2 fold insulin release by the clusters to glucose challenge. To show the scalable multiplexing for drug testing, we demonstrated the loss of stimulation index after long-term exposure to logarithmic concentration range of glybenclamide. The MPS cultured eBCs also confirmed a glycolytic bottleneck as inferred by insulin secretion responses to metabolites methyl succinate and glyceric acid. Thus, we present an innovative culture platform for eBCs with a balance of high-content and high-throughput characteristics.
Collapse
Affiliation(s)
- Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
- Department of Materials Science and Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Eleonora de Klerk
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Phichitpol Carnese
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthias Hebrok
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
- Department of Materials Science and Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
47
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
48
|
Microfluidic Technology for Evaluating and Preserving Islet Function for Islet Transplant in Type 1 Diabetes. CURRENT TRANSPLANTATION REPORTS 2022. [DOI: 10.1007/s40472-022-00377-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
49
|
Yin J, Meng H, Lin J, Ji W, Xu T, Liu H. Pancreatic islet organoids-on-a-chip: how far have we gone? J Nanobiotechnology 2022; 20:308. [PMID: 35764957 PMCID: PMC9238112 DOI: 10.1186/s12951-022-01518-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 01/10/2023] Open
Abstract
Diabetes mellitus (DM) is a disease caused by dysfunction or disruption of pancreatic islets. The advent and development of microfluidic organoids-on-a-chip platforms have facilitated reproduce of complex and dynamic environment for tissue or organ development and complex disease processes. For the research and treatment of DM, the platforms have been widely used to investigate the physiology and pathophysiology of islets. In this review, we first highlight how pancreatic islet organoids-on-a-chip have improved the reproducibility of stem cell differentiation and organoid culture. We further discuss the efficiency of microfluidics in the functional evaluation of pancreatic islet organoids, such as single-islet-sensitivity detection, long-term real-time monitoring, and automatic glucose adjustment to provide relevant stimulation. Then, we present the applications of islet-on-a-chip technology in disease modeling, drug screening and cell replacement therapy. Finally, we summarize the development and challenges of islet-on-a-chip and discuss the prospects of future research.
Collapse
Affiliation(s)
- Jiaxiang Yin
- Bioland Laboratory, Guangzhou, Guangdong, China.,Guangzhou Laboratory, Guangzhou, Guangdong, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hao Meng
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | | | - Wei Ji
- Bioland Laboratory, Guangzhou, Guangdong, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Tao Xu
- Guangzhou Laboratory, Guangzhou, Guangdong, China. .,School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China.
| | - Huisheng Liu
- Bioland Laboratory, Guangzhou, Guangdong, China. .,Guangzhou Laboratory, Guangzhou, Guangdong, China. .,School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
50
|
Yoshihara E. Adapting Physiology in Functional Human Islet Organogenesis. Front Cell Dev Biol 2022; 10:854604. [PMID: 35557947 PMCID: PMC9086403 DOI: 10.3389/fcell.2022.854604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of three-dimensional (3D)-structured functional human islets is expected to be an alternative cell source for cadaveric human islet transplantation for the treatment of insulin-dependent diabetes. Human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer infinite resources for newly synthesized human islets. Recent advancements in hPSCs technology have enabled direct differentiation to human islet-like clusters, which can sense glucose and secrete insulin, and those islet clusters can ameliorate diabetes when transplanted into rodents or non-human primates (NHPs). However, the generated hPSC-derived human islet-like clusters are functionally immature compared with primary human islets. There remains a challenge to establish a technology to create fully functional human islets in vitro, which are functionally and transcriptionally indistinguishable from cadaveric human islets. Understanding the complex differentiation and maturation pathway is necessary to generate fully functional human islets for a tremendous supply of high-quality human islets with less batch-to-batch difference for millions of patients. In this review, I summarized the current progress in the generation of 3D-structured human islets from pluripotent stem cells and discussed the importance of adapting physiology for in vitro functional human islet organogenesis and possible improvements with environmental cues.
Collapse
Affiliation(s)
- Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|