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 ÎČ 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
|
Essaouiba A, Jellali R, Poulain S, Kim SH, Danoy M, Legallais C, Sakai Y, Leclerc E. Transcriptomic characterization of the synergy between human induced pluripotent stem cells-derived liver- and pancreas-on-chip coculture. Mol Cell Endocrinol 2025; 606:112582. [PMID: 40409528 DOI: 10.1016/j.mce.2025.112582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 04/25/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Interactions between the liver and pancreas are key features of the carbohydrate and lipid homeostasis in healthy and pathological patients. To investigate the crosstalk between the two organs, we have developed an organ-on-chip coculture model derived from human induced pluripotent stem cells. The presence of pancreatic-derived tissue in the culture environment contributed to increase the CYP3A4 activity, the glycogen storage, and the expression of genes related to lipids, bile acids and sterol metabolism in the liver derived tissue. Concomitantly, the presence of liver cells led to increase the C-peptide secretion in pancreas. The coculture with liver modulated the pancreatic differentiation by increasing the activity of important transcription factors (REST, MAFB, PBX1) and by downregulating several hormone encoding genes (INS, GCG, TTR). The liver also stimulated the expression of genes involved in the response to inflammation in pancreas (via TGFÎČ/SMAD pathway). In parallel we observed a pancreatic cell reorganization coupled with the activation of the cell proliferation related transcription factor (SCRT1) and the upregulation of cellular remodeling genes (FLNA, FLNB, FN1, COL4A5). Finally, the pancreatic lipid genes were also upregulated in presence of the liver tissue. Overall, our results reflect a complex synergy between both tissues. We believe that those results are an encouraging step toward the development of relevant human model using advanced organ-on-chip technology and stem cells sources.
Collapse
Affiliation(s)
- Amal Essaouiba
- Université de Technologie de CompiÚgne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 CompiÚgne Cedex, 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
- Université de Technologie de CompiÚgne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 CompiÚgne Cedex, France.
| | - Stéphane Poulain
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Soo Hyeon Kim
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Mathieu Danoy
- Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Cécile Legallais
- Université de Technologie de CompiÚgne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 CompiÚgne Cedex, 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; Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Eric Leclerc
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan.
| |
Collapse
|
3
|
Tornberg K, Grötsch W, Ritari N, Haikka S, Sukki L, Aalto-SetÀlÀ K, Pekkanen-Mattila M, Kallio P. Compartmentalized perfusion for temporal control of the chemical microenvironment of iPSC-derived cardiac cells. LAB ON A CHIP 2025. [PMID: 40370144 DOI: 10.1039/d5lc00072f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Organ-on-chip structures are predicted to have a significant influence in drug research. In these structures, perfusion can provide cells a more controllable environment to receive signaling molecules. In many current organ-on-chip applications, perfusion is used for shear stress stimulus for the cells, but it can also provide a more precise way of controlling the chemical microenvironment around the cells. In this paper, we propose an open-top organ-on-chip structure with compartment-specific perfusion to introduce stimulating molecules to cells with only minimal extra unspecific stimulus. Using numerical simulations, we show that shear stress sensed by the cells within the structure is low. We further validated the flow profile experimentally. We showed that the hiPSC-CMs accommodate to the flow environment where the shear stress is kept below 0.035 mPa. We also show that the beating rate of hiPSC-CMs increases due to the stimulation provided by chemical stimulant molecules introduced through the flow.
Collapse
Affiliation(s)
- Kaisa Tornberg
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Wolfram Grötsch
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Niina Ritari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Saara Haikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Lassi Sukki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | | | | | - Pasi Kallio
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
4
|
Wang J, Zhang H, Qu Y, Yang Y, Xu S, Ji Z, Wang Y, Zhang X, Luo Y. An eighteen-organ microphysiological system coupling a vascular network and excretion system for drug discovery. MICROSYSTEMS & NANOENGINEERING 2025; 11:89. [PMID: 40368882 PMCID: PMC12078732 DOI: 10.1038/s41378-025-00933-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 05/16/2025]
Abstract
Physiological supporting systems, such as the vascular network and excretion system, are crucial for the effective functioning of organs. This study demonstrates that when a body-on-a-chip microdevice is coupled with miniaturized physiological support systems, it can create a multi-organ microphysiological system capable of more accurately mimicking the physiological complexity of a body, thereby offering potential for preclinical drug testing. To exemplify this concept, we have developed a model system comprising 18 types of microtissues interconnected by a vascular network that replicates the in vivo blood distribution among the organs. Furthermore, this system includes an excretory system with a micro-stirrer that ensures elimination efficiency akin to in vivo conditions. Our findings indicate that this system can: (1) survive and function for almost two months; (2) achieve two-compartment pharmacokinetics of a drug; (3) investigate the dynamic relationship between the tissue distribution and toxicity of a drug; (4) establish the multimorbidity model and evaluate the effectiveness of polypharmacy, challenging tasks with traditional animal models; (5) reduce animal usage in drug evaluations. Notably, features from points (2) to (4) are capabilities not achievable by other in vitro models. The strategy proposed in this study can also be applied to the development of multi-organ microphysiological systems that mimic the physiological complexity of human organs or the entire body.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China
| | - Huixue Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China
| | - Yueyang Qu
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Suzhou Medical College, Soochow University, #199, Renai Road, Suzhou, 215127, Jiangsu Province, China
| | - Yang Yang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China
| | - Shuhui Xu
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China
| | - Zhenni Ji
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China
| | - Yuxiu Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China
| | - Xiuli Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Suzhou Medical College, Soochow University, #199, Renai Road, Suzhou, 215127, Jiangsu Province, China.
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2, Linggong Road, Dalian, 116024, Liaoning Province, China.
| |
Collapse
|
5
|
Fakhri Bafghi MS, Khoshnam Rad N, Roostaei G, Nikfar S, Abdollahi M. The reality of modeling irritable bowel syndrome: progress and challenges. Expert Opin Drug Discov 2025; 20:433-445. [PMID: 40162721 DOI: 10.1080/17460441.2025.2481264] [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: 06/03/2024] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that is often therapeutically challenging. While research has advanced our understanding of IBS pathophysiology, developing precise models to predict drug response and treatment outcomes remains a significant hurdle. AREAS COVERED This perspective provides an overview of the use of animal models alongside cutting-edge technologies used to bring drugs from bench to bedside.Furthermore, the authors examine the progress and limitations of IBS modeling. The authors further discuss the challenges of traditional animal models and gives a spotlight to the potential of innovative technologies, such as organ-on-chip systems, computational models, and artificial intelligence (AI). These approaches intend to enhance both the understanding and treatment of IBS. EXPERT OPINION Although animal models have been central to understanding IBS research, they have limitations. The future of IBS research resides in integrating organ-on-chip systems and utilizing modern technological developments, such as AI. These tools will enable the design of more effective treatment strategies and improve patients' overall well-being. To achieve this, collaboration between experts from various disciplines is essential to improve these models and guarantee their clinical application and reliability.
Collapse
Affiliation(s)
- Maryam S Fakhri Bafghi
- Department of Internal Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Khoshnam Rad
- Thoracic Research Center, Imam Khomeini Hospital Complex, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazal Roostaei
- Thoracic Research Center, Imam Khomeini Hospital Complex, Tehran, Iran
- Rasoul Akram Hospital Clinical Research Development Center, School of Medicine, Rasool Akram Medical Complex, Iran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Personalized Medicine Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Kozalak G, KoĆar A. Bone-on-a-Chip Systems for Hematological Cancers. BIOSENSORS 2025; 15:176. [PMID: 40136973 PMCID: PMC11940066 DOI: 10.3390/bios15030176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Hematological malignancies originating from blood, bone marrow, and lymph nodes include leukemia, lymphoma, and myeloma, which necessitate the use of a distinct chemotherapeutic approach. Drug resistance frequently complicates their treatment, highlighting the need for predictive tools to guide therapeutic decisions. Conventional 2D/3D cell cultures do not fully encompass in vivo criteria, and translating disease models from mice to humans proves challenging. Organ-on-a-chip technology presents an avenue to surmount genetic disparities between species, offering precise design, concurrent manipulation of various cell types, and extrapolation of data to human physiology. The development of bone-on-a-chip (BoC) systems is crucial for accurately representing the in vivo bone microenvironment, predicting drug responses for hematological cancers, mitigating drug resistance, and facilitating personalized therapeutic interventions. BoC systems for modeling hematological cancers and drug research can encompass intricate designs and integrated platforms for analyzing drug response data to simulate disease scenarios. This review provides a comprehensive examination of BoC systems applicable to modeling hematological cancers and visualizing drug responses within the intricate context of bone. It thoroughly discusses the materials pertinent to BoC systems, suitable in vitro techniques, the predictive capabilities of BoC systems in clinical settings, and their potential for commercialization.
Collapse
Affiliation(s)
- GĂŒl Kozalak
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali KoĆar
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TĂBA), Ăankaya, Ankara 06700, Turkey
| |
Collapse
|
7
|
Chauhdari T, Zaidi SA, Su J, Ding Y. Organoids meet microfluidics: recent advancements, challenges, and future of organoids-on-chip. IN VITRO MODELS 2025; 4:71-88. [PMID: 40160209 PMCID: PMC11950471 DOI: 10.1007/s44164-025-00086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
Organoids are three-dimensional, miniaturized tissue-like structures derived from either stem cells or primary cells, emerging as powerful in vitro models for studying developmental biology, disease pathology, and drug discovery. These organoids more accurately mimic cell-cell interactions and complexities of human tissues compared to traditional cell cultures. However, challenges such as limited nutrient supply and biomechanical cue replication hinder their maturation and viability. Microfluidic technologies, with their ability to control fluid flow and mimic the mechanical environment of tissues, have been integrated with organoids to create organoid-on-chip models that address these limitations. These models not only improve the physiological relevance of organoids but also enable more precise investigation of disease mechanisms and therapeutic responses. By combining microfluidics and organoids, several advanced organoids-on-chip models have been developed to investigate mechanical and biochemical cues involved in disease progression. This review discusses various methods to develop organoids-on-chip and the recently established organoids-on-chip models with their advanced functions. Finally, we highlighted potential strategies to enhance the functionality of organoid models, aiming to overcome current limitations and bridge the gap between current cell culture models and clinical applications, advancing personalized medicine, and improving therapeutic testing.
Collapse
Affiliation(s)
- Talha Chauhdari
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| | - Syeda Armana Zaidi
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| | - Jilei Su
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| |
Collapse
|
8
|
Schellberg BG, Koppes RA, Koppes AN. Recent Advances in Integrated Organ-Chip Sensing Toward Robust and User-Friendly Systems. J Biomed Mater Res A 2025; 113:e37876. [PMID: 39893559 DOI: 10.1002/jbm.a.37876] [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: 10/18/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
Organs-on-a-chip (OOC) are an emergent technology that bridge the gap between current in vitro and in vivo models used to inform drug discovery and investigate disease pathophysiology. These systems offer improved bio-relevance and controlled complexity through the integration of physical and/or chemical stimuli matched to physiologically relevant conditions. Although significant advancements have been made toward recreating organ-specific physiology on chip, the methods available to study structure and function of the cell microenvironment are still limited. Established analysis approaches, including fluorescence microscopy, rely on laborious offline workflows that yield limited time-point data. As the OOC field continues to evolve, there is a unique opportunity to engineer improved characterization methods into organ-chip devices. This review provides an overview of current integrated sensing approaches that address current limitations and enable real-time readout of relevant physiological parameters in OOC.
Collapse
Affiliation(s)
- Bryan G Schellberg
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Hellen DJ, Ungerleider J, Tevonian E, Sphabmixay P, Roy P, Lewis C, Jeppesen J, Demozay D, Griffith LG. A Microphysiological Model of Progressive Human Hepatic Insulin Resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631261. [PMID: 39829839 PMCID: PMC11741310 DOI: 10.1101/2025.01.08.631261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background & Aims Hepatic insulin resistance is a fundamental phenomenon observed in both Type 2 diabetes (T2D) and metabolic (dysfunction) associated fatty liver disease (MAFLD). The relative contributions of nutrients, hyperinsulinemia, hormones, inflammation, and other cues are difficult to parse in vivo as they are convoluted by interplay between the local and systemic events. Here, we used a well-established human liver microphysiological system (MPS) to establish a physiologically-relevant insulin-responsive metabolic baseline and probe how primary human hepatocytes respond to controlled perturbations in insulin, glucose, and free fatty acids (FFAs). Methods Replicate liver MPS were maintained in media with either 200 pM (normal) or 800 pM (T2D) insulin for up to 3 weeks. Conditions of standard glucose (5.5 mM), hyperglycemia (11 mM glucose), normal (20ÎŒM) and elevated FFA (100 ÎŒM), alone and in combination were used at each insulin concentration, either continuously or reversing back to standard media after 2 weeks of simulated T2D conditions. Hepatic glucose production, activation of signaling pathways, insulin clearance, transcriptome analysis, and intracellular lipid and bile acid accumulation were assessed. Results Hyperinsulinemia alone induces insulin resistance after one week of exposure, while hyperglycemia and increased FFAs significantly exacerbate this phenotype. Hyperinsulinemia, along with elevated glucose and FFAs, transcriptionally predisposes hepatocytes to insulin resistance through altered metabolic and immune signaling pathways. The phenotypes observed in hyperinsulinemia and nutrient overload are partially reversible upon return to normophysiologic conditions. Conclusions Our enhanced in vitro model, replicating multiple aspects of the insulin-resistant condition, offers improved insights into disease mechanisms with relevance to human physiology.
Collapse
Affiliation(s)
- Dominick J. Hellen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139 US
| | - Jessica Ungerleider
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139 US
| | - Erin Tevonian
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139 US
| | - Pierre Sphabmixay
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139 US
| | - Priyatanu Roy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139 US
| | - Caroline Lewis
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02139 US
| | - Jacob Jeppesen
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02139 US
- Liver Disease, Novo Nordisk A/S, MÄlÞv, Denmark
| | | | - Linda G. Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139 US
| |
Collapse
|
10
|
Yoneyama Y, Wu Y, Mori K, Takebe T. In toto biological framework: Modeling interconnectedness during human development. Dev Cell 2025; 60:8-20. [PMID: 39765224 DOI: 10.1016/j.devcel.2024.09.027] [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: 08/30/2023] [Revised: 04/21/2024] [Accepted: 09/20/2024] [Indexed: 05/24/2025]
Abstract
Recent advancements in pluripotent stem cell and synthetic tissue technology have brought significant breakthroughs in studying early embryonic development, particularly within the first trimester of development in humans. However, during fetal stage development, investigating further biological events represents a major challenge, partly due to the evolving complexity and continued interaction across multiple organ systems. To bridge this gap, we propose an "in toto" biological framework that leverages a triad of technologies: synthetic tissues, intravital microscopy, and computer vision to capture in vivo cellular morphodynamics, conceptualized as single-cell choreography. This perspective will discuss the inherent challenges in capturing such complexities and explore engineering technologies to delve into the less-explored phase of human development. We also propose reframing the organ-centric to a system-centric paradigm, as such a framework broadens the value of the in vivo-embedded synthetic-tissue-based approach for interrogating the multifaceted interplay of human developmental processes during this crucial stage.
Collapse
Affiliation(s)
- Yosuke Yoneyama
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Graduate School of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan; Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yunheng Wu
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kensaku Mori
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Information Technology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Research Center for Medical Bigdata, National Institute of Informatics, Tokyo 100-0003, Japan
| | - Takanori Takebe
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Graduate School of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan; Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, and Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|
11
|
Feng X, Zhang H, Yang S, Cui D, Wu Y, Qi X, Su Z. From stem cells to pancreatic ÎČ-cells: strategies, applications, and potential treatments for diabetes. Mol Cell Biochem 2025; 480:173-190. [PMID: 38642274 DOI: 10.1007/s11010-024-04999-x] [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: 01/16/2024] [Accepted: 03/21/2024] [Indexed: 04/22/2024]
Abstract
Loss and functional failure of pancreatic ÎČ-cells results in disruption of glucose homeostasis and progression of diabetes. Although whole pancreas or pancreatic islet transplantation serves as a promising approach for ÎČ-cell replenishment and diabetes therapy, the severe scarcity of donor islets makes it unattainable for most diabetic patients. Stem cells, particularly induced pluripotent stem cells (iPSCs), are promising for the treatment of diabetes owing to their self-renewal capacity and ability to differentiate into functional ÎČ-cells. In this review, we first introduce the development of functional ÎČ-cells and their heterogeneity and then turn to highlight recent advances in the generation of ÎČ-cells from stem cells and their potential applications in disease modeling, drug discovery and clinical therapy. Finally, we have discussed the current challenges in developing stem cell-based therapeutic strategies for improving the treatment of diabetes. Although some significant technical hurdles remain, stem cells offer great hope for patients with diabetes and will certainly transform future clinical practice.
Collapse
Affiliation(s)
- Xingrong Feng
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Hongmei Zhang
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Shanshan Yang
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Daxin Cui
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Yanting Wu
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Xiaocun Qi
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Zhiguang Su
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China.
| |
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
|
Son KH, Kim DH, Park S, Kim HJ, Park M, Kim SJ, Lee SJ, Ahn K, Lee JW. Spherical Shell Bioprinting to Produce Uniform Spheroids with Controlled Sizes. J Funct Biomater 2024; 15:350. [PMID: 39590553 PMCID: PMC11595458 DOI: 10.3390/jfb15110350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Conventional cell spheroid production methods are largely manual, leading to variations in size and shape that compromise consistency and reliability for use in cell-based therapeutic applications. To enhance spheroid production, a spherical shell bioprinting system was implemented, enabling the high-throughput generation of uniform cell spheroids with precisely controlled sizes. The system encapsulates cells within thin alginate hydrogel shells formed through bioprinting and ion crosslinking reactions. Alginate-calcium ion crosslinking created alginate shells that contained gelatin-based bioinks with embedded cells, facilitating spontaneous cell aggregation within the shells and eliminating the need for plastic wells. By adjusting cell concentrations in the alginate-gelatin bioink, we achieved precise control over spheroid size, maintaining a sphericity above 0.94 and size deviations within ±10 ”m. This method has been successfully applied to various cell types including cancer cells, fibroblasts, chondrocytes, and epithelial cells, demonstrating its versatility. This scalable approach enhances the reliability of cell therapy and drug screening, offering a robust platform for future biomedical applications.
Collapse
Affiliation(s)
- Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, College of Medicine, Gachon University, 21, Namdong-daero 774 Beon-gil, Namdong-gu, Incheon 21565, Republic of Korea;
| | - Dong-Ha Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seunghye Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
| | - Hyun Jae Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Mira Park
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seung-Jin Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Keunsun Ahn
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| |
Collapse
|
14
|
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
|
15
|
Chavoshinezhad N, Niknafs B. Innovations in 3D ovarian and follicle engineering for fertility preservation and restoration. Mol Biol Rep 2024; 51:1004. [PMID: 39305382 DOI: 10.1007/s11033-024-09783-0] [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: 03/25/2024] [Accepted: 07/05/2024] [Indexed: 02/06/2025]
Abstract
In-vitro maturation (IVM) is the process of cultivating early-stage follicles from the primordial to the antral stage and facilitating the maturation of oocytes outside the body within a supportive environment. This intricate procedure requires the careful coordination of various factors to replicate the natural ovarian conditions. Advanced techniques for IVM are designed to mimic the natural ovarian environment and enhance the development of follicles. Three-dimensional (3D) culture systems provide a more biologically relevant setting for follicle growth compared to traditional two-dimensional (2D) cultures. Traditional culture systems, often fail to support the complex process of follicle development effectively. However, modern engineered reproductive tissues and culture systems are making it possible to create increasingly physiological in-vitro models of folliculogenesis. These innovative methods are enabling researchers and clinicians to better replicate the dynamic and supportive environment of the ovary, thereby improving the outcomes of IVM offering new hope for fertility preservation and treatment. This paper focuses on the routine 3D culture, and innovative 3D culture of ovary and follicles, including a tissue engineering scaffolds, microfluidic (dynamic) culture system, organ-on-chip models, EVATAR system, from a clinical perspective to determine the most effective approach for achieving in-vitro maturation of follicles. These techniques provide critical support for ovarian function in various ovarian-associated disorders, including primary ovarian insufficiency (POI), premature ovarian failure (POF), ovarian cancer, and age-related infertility.
Collapse
Affiliation(s)
- Negin Chavoshinezhad
- Immunology research center , Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomy, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrooz Niknafs
- Immunology research center , Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Anatomy, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Arjmandi H, Kanebratt KP, Vilén L, Gennemark P, Noel A. 3D cell aggregates amplify diffusion signals. PLoS One 2024; 19:e0310109. [PMID: 39264935 DOI: 10.1371/journal.pone.0310109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
Biophysical models can predict the behavior of cell cultures including 3D cell aggregates (3DCAs), thereby reducing the need for costly and time-consuming experiments. Specifically, mass transfer models enable studying the transport of nutrients, oxygen, signaling molecules, and drugs in 3DCA. These models require the defining of boundary conditions (BC) between the 3DCA and surrounding medium. However, accurately modeling the BC that relates the inner and outer boundary concentrations at the border between the 3DCA and the medium remains a challenge that this paper addresses using both theoretical and experimental methods. The provided biophysical analysis indicates that the concentration of molecules inside boundary is higher than that at the outer boundary, revealing an amplification factor that is confirmed by a particle-based simulator (PBS). Due to the amplification factor, the PBS confirms that when a 3DCA with a low concentration of target molecules is introduced to a culture medium with a higher concentration, the molecule concentration in the medium rapidly decreases. The theoretical model and PBS simulations were used to design a pilot experiment with liver spheroids as the 3DCA and glucose as the target molecule. Experimental results agree with the proposed theory and derived properties.
Collapse
Affiliation(s)
- Hamidreza Arjmandi
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Kajsa P Kanebratt
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
| | - Liisa Vilén
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
| | - Peter Gennemark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Adam Noel
- School of Engineering, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
17
|
Hosseinzadeh M, Saber N, Bidar SS, Hashemi S, Teymoori F, Mirzaei M, Nadjarzadeh A, Rahideh ST. Association of dietary and lifestyle inflammatory indices with type 2 diabetes risk in Iranian adults. BMC Endocr Disord 2024; 24:131. [PMID: 39085857 PMCID: PMC11292998 DOI: 10.1186/s12902-024-01673-7] [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: 01/17/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is often linked to chronic inflammation, which can be influenced by both lifestyle and dietary choices. However, the relationship between the inflammatory potential of diet and lifestyle factors and the risk of developing T2DM remains unclear. The present study aimed to investigate the associations of the empirical dietary inflammatory index (EDII), dietary inflammatory score (DIS), and lifestyle inflammatory score (LIS) with the risk of T2DM among Iranian adults. METHODS The current study was conducted on 5714 individuals from Yazd Health Study (YaHS) who were followed up for a mean period of six years. YaHS is a prospective cohort study which has been conducted since 2014. Dietary intakes were collected at baseline using the food frequency questionnaire. The relative risk (RR) of T2DM was calculated by Cox regression analysis across tertiles of EDII, DIS, and LIS, adjusted for potential confounders. RESULTS The meanâ±âSD for the age and body mass index of the study population were 47.0â±â9.2 years and 26.7â±â5.1 Kg.m2, respectively. A significant association between LIS and the risk of T2DM was observed (RR: 4.05, 95% CI: 2.61-6.27 P-trendâ<â0.001). Individuals in the highest compared to the lowest tertile of EDII-LIS (RR: 3.07, 95%CI: 2.01-4.68; P for trendâ<â0.001) and DIS-LIS (RR: 2.42, 95%CI: 1.69-3.49; P for trendâ<â0.001) had a higher risk of T2DM. However, no significant association was found between EDII and DIS scores and the risk of T2DM. CONCLUSION Greater adherence to LIS, EDII-LIS, and DIS-LIS scores was associated with a higher risk of T2DM, while no significant association was found between EDII and DIS with T2DM risk.
Collapse
Affiliation(s)
- Mahdieh Hosseinzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Niloufar Saber
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sakineh Shab Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sogol Hashemi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farshad Teymoori
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Mirzaei
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azadeh Nadjarzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyedeh Tayebeh Rahideh
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Rigal S, Casas B, Kanebratt KP, Wennberg Huldt C, Magnusson LU, MĂŒllers E, Karlsson F, Clausen M, Hansson SF, Leonard L, Cairns J, Jansson Löfmark R, ĂmmĂ€lĂ€ C, Marx U, Gennemark P, Cedersund G, Andersson TB, VilĂ©n LK. Normoglycemia and physiological cortisone level maintain glucose homeostasis in a pancreas-liver microphysiological system. Commun Biol 2024; 7:877. [PMID: 39025915 PMCID: PMC11258270 DOI: 10.1038/s42003-024-06514-w] [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: 06/18/2023] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Current research on metabolic disorders and diabetes relies on animal models because multi-organ diseases cannot be well studied with standard in vitro assays. Here, we have connected cell models of key metabolic organs, the pancreas and liver, on a microfluidic chip to enable diabetes research in a human-based in vitro system. Aided by mechanistic mathematical modeling, we demonstrate that hyperglycemia and high cortisone concentration induce glucose dysregulation in the pancreas-liver microphysiological system (MPS), mimicking a diabetic phenotype seen in patients with glucocorticoid-induced diabetes. In this diseased condition, the pancreas-liver MPS displays beta-cell dysfunction, steatosis, elevated ketone-body secretion, increased glycogen storage, and upregulated gluconeogenic gene expression. Conversely, a physiological culture condition maintains glucose tolerance and beta-cell function. This method was reproducible in two laboratories and was effective in multiple pancreatic islet donors. The model also provides a platform to identify new therapeutic proteins, as demonstrated with a combined transcriptome and proteome analysis.
Collapse
Affiliation(s)
| | - Belén Casas
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Kajsa P Kanebratt
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Charlotte Wennberg Huldt
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lisa U Magnusson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Erik MĂŒllers
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Fredrik Karlsson
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Maryam Clausen
- Translational Genomics, Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Sara F Hansson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Louise Leonard
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Jonathan Cairns
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Rasmus Jansson Löfmark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Carina ĂmmĂ€lĂ€
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Peter Gennemark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Gunnar Cedersund
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Tommy B Andersson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Liisa K Vilén
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
19
|
Jadalannagari S, Ewart L. Beyond the hype and toward application: liver complex in vitro models in preclinical drug safety. Expert Opin Drug Metab Toxicol 2024; 20:607-619. [PMID: 38465923 DOI: 10.1080/17425255.2024.2328794] [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: 01/15/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Drug induced Liver-Injury (DILI) is a leading cause of drug attrition and complex in vitro models (CIVMs), including three dimensional (3D) spheroids, 3D bio printed tissues and flow-based systems, could improve preclinical prediction. Although CIVMs have demonstrated good sensitivity and specificity in DILI detection their adoption remains limited. AREAS COVERED This article describes DILI, the challenges with its prediction and the current strategies and models that are being used. It reviews data from industry-FDA collaborations and strategic partnerships and finishes with an outlook of CIVMs in preclinical toxicity testing. Literature searches were performed using PubMed and Google Scholar while product information was collected from manufacturer websites. EXPERT OPINION Liver CIVMs are promising models for predicting DILI although, a decade after their introduction, routine use by the pharmaceutical industry is limited. To accelerate their adoption, several industry-regulator-developer partnerships or consortia have been established to guide the development and qualification. Beyond this, liver CIVMs should continue evolving to capture greater immunological mimicry while partnering with computational approaches to deliver systems that change the paradigm of predicting DILI.
Collapse
Affiliation(s)
| | - Lorna Ewart
- Department of Bioinnovations, Emulate Inc, Boston, MA, USA
| |
Collapse
|
20
|
Thorne D, McHugh D, Simms L, Lee KM, Fujimoto H, Moses S, Gaca M. Applying new approach methodologies to assess next-generation tobacco and nicotine products. FRONTIERS IN TOXICOLOGY 2024; 6:1376118. [PMID: 38938663 PMCID: PMC11208635 DOI: 10.3389/ftox.2024.1376118] [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: 01/25/2024] [Accepted: 04/30/2024] [Indexed: 06/29/2024] Open
Abstract
In vitro toxicology research has accelerated with the use of in silico, computational approaches and human in vitro tissue systems, facilitating major improvements evaluating the safety and health risks of novel consumer products. Innovation in molecular and cellular biology has shifted testing paradigms, with less reliance on low-throughput animal data and greater use of medium- and high-throughput in vitro cellular screening approaches. These new approach methodologies (NAMs) are being implemented in other industry sectors for chemical testing, screening candidate drugs and prototype consumer products, driven by the need for reliable, human-relevant approaches. Routine toxicological methods are largely unchanged since development over 50 years ago, using high-doses and often employing in vivo testing. Several disadvantages are encountered conducting or extrapolating data from animal studies due to differences in metabolism or exposure. The last decade saw considerable advancement in the development of in vitro tools and capabilities, and the challenges of the next decade will be integrating these platforms into applied product testing and acceptance by regulatory bodies. Governmental and validation agencies have launched and applied frameworks and "roadmaps" to support agile validation and acceptance of NAMs. Next-generation tobacco and nicotine products (NGPs) have the potential to offer reduced risks to smokers compared to cigarettes. These include heated tobacco products (HTPs) that heat but do not burn tobacco; vapor products also termed electronic nicotine delivery systems (ENDS), that heat an e-liquid to produce an inhalable aerosol; oral smokeless tobacco products (e.g., Swedish-style snus) and tobacco-free oral nicotine pouches. With the increased availability of NGPs and the requirement of scientific studies to support regulatory approval, NAMs approaches can supplement the assessment of NGPs. This review explores how NAMs can be applied to assess NGPs, highlighting key considerations, including the use of appropriate in vitro model systems, deploying screening approaches for hazard identification, and the importance of test article characterization. The importance and opportunity for fit-for-purpose testing and method standardization are discussed, highlighting the value of industry and cross-industry collaborations. Supporting the development of methods that are accepted by regulatory bodies could lead to the implementation of NAMs for tobacco and nicotine NGP testing.
Collapse
Affiliation(s)
- David Thorne
- BAT (Investments) Ltd., Southampton, Hampshire, United Kingdom
| | - Damian McHugh
- PMI R&D Philip Morris Products S. A., NeuchĂątel, Switzerland
| | - Liam Simms
- Imperial Brands, Bristol, United Kingdom
| | - K. Monica Lee
- Altria Client Services LLC, Richmond, VA, United States
| | | | | | - Marianna Gaca
- BAT (Investments) Ltd., Southampton, Hampshire, United Kingdom
| |
Collapse
|
21
|
Wang C, Abadpour S, Aizenshtadt A, Dalmao-Fernandez A, HĂžyem M, Wilhelmsen I, Stokowiec J, Olsen PA, Krauss S, Chera S, Ghila L, RĂŠder H, Scholz H. Cell identity dynamics and insight into insulin secretagogues when employing stem cell-derived islets for disease modeling. Front Bioeng Biotechnol 2024; 12:1392575. [PMID: 38933536 PMCID: PMC11199790 DOI: 10.3389/fbioe.2024.1392575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Stem cell-derived islets (SC-islets) are not only an unlimited source for cell-based therapy of type 1 diabetes but have also emerged as an attractive material for modeling diabetes and conducting screening for treatment options. Prior to SC-islets becoming the established standard for disease modeling and drug development, it is essential to understand their response to various nutrient sources in vitro. This study demonstrates an enhanced efficiency of pancreatic endocrine cell differentiation through the incorporation of WNT signaling inhibition following the definitive endoderm stage. We have identified a tri-hormonal cell population within SC-islets, which undergoes reduction concurrent with the emergence of elevated numbers of glucagon-positive cells during extended in vitro culture. Over a 6-week period of in vitro culture, the SC-islets consistently demonstrated robust insulin secretion in response to glucose stimulation. Moreover, they manifested diverse reactivity patterns when exposed to distinct nutrient sources and exhibited deviant glycolytic metabolic characteristics in comparison to human primary islets. Although the SC-islets demonstrated an aberrant glucose metabolism trafficking, the evaluation of a potential antidiabetic drug, pyruvate kinase agonist known as TEPP46, significantly improved in vitro insulin secretion of SC-islets. Overall, this study provided cell identity dynamics investigation of SC-islets during prolonged culturing in vitro, and insights into insulin secretagogues. Associated advantages and limitations were discussed when employing SC-islets for disease modeling.
Collapse
Affiliation(s)
- Chencheng Wang
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
| | - Shadab Abadpour
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
| | | | - Andrea Dalmao-Fernandez
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Merete HĂžyem
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | - Ingrid Wilhelmsen
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Justyna Stokowiec
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
| | - Petter Angell Olsen
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Stefan Krauss
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Simona Chera
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Luiza Ghila
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Helge RĂŠder
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Hanne Scholz
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub, Center of Excellence, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
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 MedicineCedarsâSinai Medical CenterLos AngelesCaliforniaUSA
| | - Ling Li
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
23
|
Chen J, Lu J, Wang SN, Miao CY. Application and challenge of pancreatic organoids in therapeutic research. Front Pharmacol 2024; 15:1366417. [PMID: 38855754 PMCID: PMC11157021 DOI: 10.3389/fphar.2024.1366417] [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: 01/06/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
The in-vivo non-human primate animal and in-vitro cell disease models play a crucial part in the study of the mechanisms underlying the occurrence and development of pancreatic diseases, but with increasingly prominent limitations with in-depth research. Organoids derived from human pluripotent and adult stem cells resemble human in-vivo organs in their cellular composition, spatial tissue structure and physiological function, making them as an advantageous research tool. Up until now, numerous human organoids, including pancreas, have been effectively developed, demonstrating significant potential for research in organ development, disease modeling, drug screening, and regenerative medicine. However, different from intestine, liver and other organs, the pancreas is the only special organ in the human body, consisting of an exocrine gland and an endocrine gland. Thus, the development of pancreatic organoid technology faces greater challenges, and how to construct a composite pancreatic organoid with exocrine and endocrine gland is still difficult in current research. By reviewing the fundamental architecture and physiological role of the human pancreas, along with the swiftly developing domain of pancreatic organoids, we summarize the method and characteristics of human pancreatic organoids, and its application in modeling pancreatic diseases, as a platform for individualized drug screening and in regenerative medicine study. As the first comprehensive review that focus on the pharmacological study of human pancreatic organoid, the review hopes to help scholars to have a deeper understanding in the study of pancreatic organoid.
Collapse
Affiliation(s)
- Jin Chen
- Department of Endocrinology and Metabolism, Changhai Hospital, Second Military University /Naval Medical University, Shanghai, China
- Department of Pharmacology, Second Military Medical University /Naval Medical University, Shanghai, China
| | - Jin Lu
- Department of Endocrinology and Metabolism, Changhai Hospital, Second Military University /Naval Medical University, Shanghai, China
| | - Shu-Na Wang
- Department of Pharmacology, Second Military Medical University /Naval Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University /Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Ingelman-Sundberg M, Lauschke VM. Individualized Pharmacotherapy Utilizing Genetic Biomarkers and Novel In Vitro Systems As Predictive Tools for Optimal Drug Development and Treatment. Drug Metab Dispos 2024; 52:467-475. [PMID: 38575185 DOI: 10.1124/dmd.123.001302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/15/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
In the area of drug development and clinical pharmacotherapy, a profound understanding of the pharmacokinetics and potential adverse reactions associated with the drug under investigation is paramount. Essential to this endeavor is a comprehensive understanding about interindividual variations in absorption, distribution, metabolism, and excretion (ADME) genetics and the predictive capabilities of in vitro systems, shedding light on metabolite formation and the risk of adverse drug reactions (ADRs). Both the domains of pharmacogenomics and the advancement of in vitro systems are experiencing rapid expansion. Here we present an update on these burgeoning fields, providing an overview of their current status and illuminating potential future directions. SIGNIFICANCE STATEMENT: There is very rapid development in the area of pharmacogenomics and in vitro systems for predicting drug pharmacokinetics and risk for adverse drug reactions. We provide an update of the current status of pharmacogenomics and developed in vitro systems on these aspects aimed to achieve a better personalized pharmacotherapy.
Collapse
Affiliation(s)
- Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.I.-S., V.M.L.); Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of TĂŒbingen, TĂŒbingen, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.I.-S., V.M.L.); Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of TĂŒbingen, TĂŒbingen, Germany (V.M.L.)
| |
Collapse
|
25
|
Horn G, Worek F. Suitability of human HepaRG cells and liver spheroids as in vitro model to investigate the bioactivation of the organothiophosphate pesticide parathion. Toxicol In Vitro 2024; 97:105811. [PMID: 38521251 DOI: 10.1016/j.tiv.2024.105811] [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: 10/24/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 03/25/2024]
Abstract
Organophosphorus compounds (OP) constitute a large group of chemicals including pesticides and nerve agents. Organothiophosphate pesticides require cytochrome P450-mediated oxidative desulphuration in the liver to form corresponding oxons, which are potent inhibitors of the enzyme acetylcholinesterase (AChE). Human HepaRG cells are a promising tool to study liver-specific functions and have been shown to maintain drug metabolizing enzymes. This research describes for the first time the in vitro metabolic activation of an organothiophosphate to its active oxon by two different HepaRG cell-based models. Monolayer cultures and liver spheroids were exposed to the model OP parathion and the quantification of the corresponding oxon was performed with an AChE inhibition assay. Our results showed a time- and dose-dependent cytochrome P450 catalyzed bioactivation and a superior metabolism capacity of the monolayer HepaRG model in comparison with the liver spheroids. Finally, HepaRG cells can be assessed as a metabolically competent cell model intermediate between cell-free preparations and intact animals and as suitable to study OP metabolism in the human liver.
Collapse
Affiliation(s)
- Gabriele Horn
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany.
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| |
Collapse
|
26
|
Aizenshtadt A, Wang C, Abadpour S, Menezes PD, Wilhelmsen I, DalmaoâFernandez A, Stokowiec J, Golovin A, Johnsen M, Combriat TMD, RĂžbergâLarsen H, Gadegaard N, Scholz H, Busek M, Krauss SJK. Pump-Less, Recirculating Organ-on-Chip (rOoC) Platform to Model the Metabolic Crosstalk between Islets and Liver. Adv Healthc Mater 2024; 13:e2303785. [PMID: 38221504 PMCID: PMC11468483 DOI: 10.1002/adhm.202303785] [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: 10/31/2023] [Revised: 12/05/2023] [Indexed: 01/16/2024]
Abstract
Type 2 diabetes mellitus (T2DM), obesity, and metabolic dysfunction-associated steatotic liver disease (MASLD) are epidemiologically correlated disorders with a worldwide growing prevalence. While the mechanisms leading to the onset and development of these conditions are not fully understood, predictive tissue representations for studying the coordinated interactions between central organs that regulate energy metabolism, particularly the liver and pancreatic islets, are needed. Here, a dual pump-less recirculating organ-on-chip platform that combines human pluripotent stem cell (sc)-derived sc-liver and sc-islet organoids is presented. The platform reproduces key aspects of the metabolic cross-talk between both organs, including glucose levels and selected hormones, and supports the viability and functionality of both sc-islet and sc-liver organoids while preserving a reduced release of pro-inflammatory cytokines. In a model of metabolic disruption in response to treatment with high lipids and fructose, sc-liver organoids exhibit hallmarks of steatosis and insulin resistance, while sc-islets produce pro-inflammatory cytokines on-chip. Finally, the platform reproduces known effects of anti-diabetic drugs on-chip. Taken together, the platform provides a basis for functional studies of obesity, T2DM, and MASLD on-chip, as well as for testing potential therapeutic interventions.
Collapse
Affiliation(s)
- Aleksandra Aizenshtadt
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Chencheng Wang
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Shadab Abadpour
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
- Institute for Surgical ResearchOslo University HospitalOsloNorway
| | - Pedro Duarte Menezes
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Ingrid Wilhelmsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Andrea DalmaoâFernandez
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Department of PharmacyFaculty of Mathematics and Natural SciencesUniversity of OsloP.O. Box 1083Oslo0316Norway
| | - Justyna Stokowiec
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Alexey Golovin
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mads Johnsen
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Thomas M. D. Combriat
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
| | - Hanne RĂžbergâLarsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Nikolaj Gadegaard
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Hanne Scholz
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mathias Busek
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Stefan J. K. Krauss
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| |
Collapse
|
27
|
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
|
28
|
Ponmozhi J, Dhinakaran S, Kocsis D, IvĂĄn K, ErdĆ F. Models for barrier understanding in health and disease in lab-on-a-chips. Tissue Barriers 2024; 12:2221632. [PMID: 37294075 PMCID: PMC11042069 DOI: 10.1080/21688370.2023.2221632] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
The maintenance of body homeostasis relies heavily on physiological barriers. Dysfunction of these barriers can lead to various pathological processes, including increased exposure to toxic materials and microorganisms. Various methods exist to investigate barrier function in vivo and in vitro. To investigate barrier function in a highly reproducible manner, ethically, and high throughput, researchers have turned to non-animal techniques and micro-scale technologies. In this comprehensive review, the authors summarize the current applications of organ-on-a-chip microfluidic devices in the study of physiological barriers. The review covers the blood-brain barrier, ocular barriers, dermal barrier, respiratory barriers, intestinal, hepatobiliary, and renal/bladder barriers under both healthy and pathological conditions. The article then briefly presents placental/vaginal, and tumour/multi-organ barriers in organ-on-a-chip devices. Finally, the review discusses Computational Fluid Dynamics in microfluidic systems that integrate biological barriers. This article provides a concise yet informative overview of the current state-of-the-art in barrier studies using microfluidic devices.
Collapse
Affiliation(s)
- J. Ponmozhi
- Microfluidics Laboratory, Department of Mechanical Engineering, IPS Academy-Institute of Engineering Science, Indore, India
| | - S. Dhinakaran
- The Centre for Fluid Dynamics, Department of Mechanical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Dorottya Kocsis
- Faculty of Information Technology and Bionics, Påzmåny Péter Catholic University, Budapest, Hungary
| | - KristĂłf IvĂĄn
- Faculty of Information Technology and Bionics, Påzmåny Péter Catholic University, Budapest, Hungary
| | - Franciska ErdĆ
- Faculty of Information Technology and Bionics, Påzmåny Péter Catholic University, Budapest, Hungary
| |
Collapse
|
29
|
SchlĂŒnder 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: ÎČ-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 SchlĂŒnder
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University TĂŒbingen, TĂŒbingen, Germany.
- NMI Natural and Medical Sciences Institute at the University of TĂŒbingen, Reutlingen, Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University TĂŒbingen, TĂŒbingen, Germany.
| | - Aline Zbinden
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University TĂŒbingen, TĂŒbingen, 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 TĂŒbingen, Reutlingen, Germany
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University TĂŒbingen, TĂŒbingen, Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University TĂŒbingen, TĂŒbingen, Germany.
- NMI Natural and Medical Sciences Institute at the University of TĂŒbingen, Reutlingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University TĂŒbingen, TĂŒbingen, Germany
| |
Collapse
|
30
|
Skardal A. Grand challenges in organoid and organ-on-a-chip technologies. Front Bioeng Biotechnol 2024; 12:1366280. [PMID: 38456004 PMCID: PMC10919399 DOI: 10.3389/fbioe.2024.1366280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 03/09/2024] Open
Affiliation(s)
- Aleksander Skardal
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, United States
- Cancer Biology Program, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
31
|
Hu C, Yang S, Zhang T, Ge Y, Chen Z, Zhang J, Pu Y, Liang G. Organoids and organoids-on-a-chip as the new testing strategies for environmental toxicology-applications & advantages. ENVIRONMENT INTERNATIONAL 2024; 184:108415. [PMID: 38309193 DOI: 10.1016/j.envint.2024.108415] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 02/05/2024]
Abstract
An increasing number of harmful environmental factors are causing serious impacts on human health, and there is an urgent need to accurately identify the toxic effects and mechanisms of these harmful environmental factors. However, traditional toxicity test methods (e.g., animal models and cell lines) often fail to provide accurate results. Fortunately, organoids differentiated from stem cells can more accurately, sensitively and specifically reflect the effects of harmful environmental factors on the human body. They are also suitable for specific studies and are frequently used in environmental toxicology nowadays. As a combination of organoids and organ-on-a-chip technology, organoids-on-a-chip has great potential in environmental toxicology. It is more controllable to the physicochemical microenvironment and is not easy to be contaminated. It has higher homogeneity in the size and shape of organoids. In addition, it can achieve vascularization and exchange the nutrients and metabolic wastes in time. Multi-organoids-chip can also simulate the interactions of different organs. These advantages can facilitate better function and maturity of organoids, which can also make up for the shortcomings of common organoids to a certain extent. This review firstly discussed the limitations of traditional toxicology testing platforms, leading to the introduction of new platforms: organoids and organoids-on-a-chip. Next, the applications of different organoids and organoids-on-a-chip in environmental toxicology were summarized and prospected. Since the advantages of the new platforms have not been sufficiently considered in previous literature, we particularly emphasized them. Finally, this review also summarized the opportunities and challenges faced by organoids and organoids-on-a-chip, with the expectation that readers will gain a deeper understanding of their value in the field of environmental toxicology.
Collapse
Affiliation(s)
- Chengyu Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Tianyi Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
32
|
Zhang S, Xu G, Wu J, Liu X, Fan Y, Chen J, Wallace G, Gu Q. Microphysiological Constructs and Systems: Biofabrication Tactics, Biomimetic Evaluation Approaches, and Biomedical Applications. SMALL METHODS 2024; 8:e2300685. [PMID: 37798902 DOI: 10.1002/smtd.202300685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/23/2023] [Indexed: 10/07/2023]
Abstract
In recent decades, microphysiological constructs and systems (MPCs and MPSs) have undergone significant development, ranging from self-organized organoids to high-throughput organ-on-a-chip platforms. Advances in biomaterials, bioinks, 3D bioprinting, micro/nanofabrication, and sensor technologies have contributed to diverse and innovative biofabrication tactics. MPCs and MPSs, particularly tissue chips relevant to absorption, distribution, metabolism, excretion, and toxicity, have demonstrated potential as precise, efficient, and economical alternatives to animal models for drug discovery and personalized medicine. However, current approaches mainly focus on the in vitro recapitulation of the human anatomical structure and physiological-biochemical indices at a single or a few simple levels. This review highlights the recent remarkable progress in MPC and MPS models and their applications. The challenges that must be addressed to assess the reliability, quantify the techniques, and utilize the fidelity of the models are also discussed.
Collapse
Affiliation(s)
- Shuyu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Guoshi Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Juan Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Xiao Liu
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yong Fan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jun Chen
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Gordon Wallace
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| |
Collapse
|
33
|
Abstract
Major advances in scientific discovery and insights that stem from the development and use of new techniques and models can bring remarkable progress to conventional toxicology. Although animal testing is still considered as the "gold standard" in traditional toxicity testing, there is a necessity for shift from animal testing to alternative methods regarding the drug safety testing owing to the emerging state-of-art techniques and the proposal of 3Rs (replace, reduce, and refine) towards animal welfare. This review describes some recent research methods in drug discovery toxicology, including in vitro cell and organ-on-a-chip, imaging systems, model organisms (C. elegans, Danio rerio, and Drosophila melanogaster), and toxicogenomics in modern toxicology testing.
Collapse
Affiliation(s)
- Bowen Tang
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| | - Vijay More
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| |
Collapse
|
34
|
Moon HR, Surianarayanan N, Singh T, Han B. Microphysiological systems as reliable drug discovery and evaluation tools: Evolution from innovation to maturity. BIOMICROFLUIDICS 2023; 17:061504. [PMID: 38162229 PMCID: PMC10756708 DOI: 10.1063/5.0179444] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Microphysiological systems (MPSs), also known as organ-on-chip or disease-on-chip, have recently emerged to reconstitute the in vivo cellular microenvironment of various organs and diseases on in vitro platforms. These microfluidics-based platforms are developed to provide reliable drug discovery and regulatory evaluation testbeds. Despite recent emergences and advances of various MPS platforms, their adoption of drug discovery and evaluation processes still lags. This delay is mainly due to a lack of rigorous standards with reproducibility and reliability, and practical difficulties to be adopted in pharmaceutical research and industry settings. This review discusses the current and potential use of MPS platforms in drug discovery processes while considering the context of several key steps during drug discovery processes, including target identification and validation, preclinical evaluation, and clinical trials. Opportunities and challenges are also discussed for the broader dissemination and adoption of MPSs in various drug discovery and regulatory evaluation steps. Addressing these challenges will transform long and expensive drug discovery and evaluation processes into more efficient discovery, screening, and approval of innovative drugs.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | | | - Tarun Singh
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Bumsoo Han
- Author to whom correspondence should be addressed:. Tel: +1-765-494-5626
| |
Collapse
|
35
|
Gabbin B, Meraviglia V, Angenent ML, Ward-van Oostwaard D, Sol W, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, Bellin M. Heart and kidney organoids maintain organ-specific function in a microfluidic system. Mater Today Bio 2023; 23:100818. [PMID: 37810749 PMCID: PMC10550812 DOI: 10.1016/j.mtbio.2023.100818] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/13/2023] [Accepted: 09/23/2023] [Indexed: 10/10/2023] Open
Abstract
Heart and kidney communicate with one another in an interdependent relationship and they influence each other's behavior reciprocally, as pathological changes in one organ can damage the other. Although independent human in vitro models for heart and kidney exist, they do not capture their dynamic crosstalk. We have developed a microfluidic system which can be used to study heart and kidney interaction in vitro. Cardiac microtissues (cMTs) and kidney organoids (kOs) derived from human induced pluripotent stem cells (hiPSCs) were generated and loaded into two separated communicating chambers of a perfusion chip. Static culture conditions were compared with dynamic culture under unidirectional flow. Tissue viability was maintained for minimally 72Â h under both conditions, as indicated by the presence of sarcomeric structures coupled with beating activity in cMTs and the presence of nephron structures and albumin uptake in kOs. We concluded that this system enables the study of human cardiac and kidney organoid interaction in vitro while controlling parameters like fluidic flow speed and direction. Together, this "cardiorenal-unit" provides a new in vitro model to study the cardiorenal axis and it may be further developed to investigate diseases involving both two organs and their potential treatments.
Collapse
Affiliation(s)
- Beatrice Gabbin
- Department of Anatomy and Embryology, Leiden University Medical Center, the Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, the Netherlands
| | - Maricke L. Angenent
- Department of Anatomy and Embryology, Leiden University Medical Center, the Netherlands
| | | | - Wendy Sol
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, the Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, the Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, the Netherlands
| | - Ton J. Rabelink
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, the Netherlands
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, the Netherlands
| | - Cathelijne W. van den Berg
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, the Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
36
|
Pandey S, Chmelir T, Chottova Dvorakova M. Animal Models in Diabetic Research-History, Presence, and Future Perspectives. Biomedicines 2023; 11:2852. [PMID: 37893225 PMCID: PMC10603837 DOI: 10.3390/biomedicines11102852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetes mellitus (DM) is a very serious disease, the incidence of which has been increasing worldwide. The beginning of diabetic research can be traced back to the 17th century. Since then, animals have been experimented on for diabetic research. However, the greatest development of diabetes research occurred in the second half of the last century, along with the development of laboratory techniques. Information obtained by monitoring patients and animal models led to the finding that there are several types of DM that differ significantly from each other in the causes of the onset and course of the disease. Through different types of animal models, researchers have studied the pathophysiology of all types of diabetic conditions and discovered suitable methods for therapy. Interestingly, despite the unquestionable success in understanding DM through animal models, we did not fully succeed in transferring the data obtained from animal models to human clinical research. On the contrary, we have observed that the chances of drug failure in human clinical trials are very high. In this review, we will summarize the history and presence of animal models in the research of DM over the last hundred years. Furthermore, we have summarized the new methodological approaches, such as "organ-on-chip," that have the potential to screen the newly discovered drugs for human clinical trials and advance the level of knowledge about diabetes, as well as its therapy, towards a personalized approach.
Collapse
Affiliation(s)
- Shashank Pandey
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, 323 00 Pilsen, Czech Republic;
- Department of Pharmacology and Toxicology, Faculty of Medicine in Pilsen, Charles University, 323 00 Pilsen, Czech Republic
| | - Tomas Chmelir
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, 323 00 Pilsen, Czech Republic;
| | - Magdalena Chottova Dvorakova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, 323 00 Pilsen, Czech Republic;
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, 323 00 Pilsen, Czech Republic;
| |
Collapse
|
37
|
Lenhart A, Kennedy RT. Evaluation of Surface Treatments of PDMS Microfluidic Devices for Improving Small-Molecule Recovery with Application to Monitoring Metabolites Secreted from Islets of Langerhans. ACS MEASUREMENT SCIENCE AU 2023; 3:380-389. [PMID: 37868359 PMCID: PMC10588933 DOI: 10.1021/acsmeasuresciau.3c00025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 10/24/2023]
Abstract
Microfluidic devices are becoming an important tool for bioanalysis with applications including studying cell secretion, cell growth, and drug delivery. Small molecules such as drugs, cell products, or nutrients may partition into polydimethylsiloxane (PDMS), a commonly used material for microfluidic devices, potentially leading to poor recovery or inaccurate delivery of such chemicals. To decrease small-molecule partitioning, surface and bulk PDMS treatments have been developed; however, these have been tested on few analytes, or their biocompatibility are unknown. Studies often focus on one analyte, whereas a diversity of chemicals are of interest and possibly affected. In this study, 11 device treatments are tested and applied to 21 biologically relevant small molecules with a variety of chemical structures. Device treatments are characterized using water contact angle measurements and evaluated by measuring recovery of the 21 target analytes using liquid chromatography-mass spectrometry. 1,5-Dimethyl-1,5-diazaundecamethylene polymethobromide (polybrene), a positively charged polymer, produced the least hydrophilic surface and was found to provide the best recovery with most of the analytes having >50% recovery and up to 92% recovery; however, recovery varied by analyte highlighting the importance of analyte diversity rather than targeting a single analyte in evaluating treatments. A polybrene-treated device was applied to investigate secretion from pancreatic islets, which are micro-organs involved in glucose homeostasis and diabetes. Islets secrete small molecules that have been shown to modulate the secretion of islets' main functional products, glucose-regulating hormones. The polybrene treatment enabled the detection of 20 target analytes from islets-on-chip during isosmotic and hypo-osmotic glucose perfusions and resulted in detection of more significant secretion changes compared to untreated PDMS.
Collapse
Affiliation(s)
- Ashley
E. Lenhart
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
38
|
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 ÎČ-cell health and function ex vivo is a fundamental aspect of diabetes research, including mechanistic studies, evaluation of ÎČ-cell health modulators, and development and quality control of replacement ÎČ-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 ÎČ-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 ÎČ-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 ÎČ-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
|
39
|
Fuchs S, Rieger V, Tjell AĂ, Spitz S, Brandauer K, Schaller-Ammann R, Feiel J, Ertl P, Klimant I, Mayr T. Optical glucose sensor for microfluidic cell culture systems. Biosens Bioelectron 2023; 237:115491. [PMID: 37413826 DOI: 10.1016/j.bios.2023.115491] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Glucose is the primary energy source of human cells. Therefore, monitoring glucose inside microphysiological systems (MPS) provides valuable information on the viability and metabolic state of the cultured cells. However, continuous glucose monitoring inside MPS is challenging due to a lack of suitable miniaturized sensors. Here we present an enzymatic, optical glucose sensor element for measurement inside microfluidic systems. The miniaturized glucose sensor (à 1 mm) is fabricated together with a reference oxygen sensor onto biocompatible, pressure-sensitive adhesive tape for easy integration inside microfluidic systems. Furthermore, the proposed microfluidic system can be used as plug and play sensor system with existing MPS. It was characterized under cell culture conditions (37 °C and pH 7.4) for five days, exhibiting minor drift (3% day-1). The influence of further cell culture parameters like oxygen concentration, pH, flow rate, and sterilization methods was investigated. The plug-and-play system was used for at-line measurements of glucose levels in (static) cell culture and achieved good agreement with a commercially available glucose sensor. In conclusion, we developed an optical glucose sensor element that can be easily integrated in microfluidic systems and is able to perform stable glucose measurements under cell culture conditions.
Collapse
Affiliation(s)
- Stefanie Fuchs
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9/II, 8010, Graz, Austria
| | - Veronika Rieger
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9/II, 8010, Graz, Austria
| | - Anders Ă Tjell
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9/II, 8010, Graz, Austria
| | - Sarah Spitz
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9/163-164, 1060, Vienna, Austria
| | - Konstanze Brandauer
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9/163-164, 1060, Vienna, Austria
| | - Roland Schaller-Ammann
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft m.b.H, Neue StiftingtalstraĂe 2, 8010, Graz, Austria
| | - JĂŒrgen Feiel
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft m.b.H, Neue StiftingtalstraĂe 2, 8010, Graz, Austria
| | - Peter Ertl
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9/163-164, 1060, Vienna, Austria
| | - Ingo Klimant
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9/II, 8010, Graz, Austria
| | - Torsten Mayr
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9/II, 8010, Graz, Austria.
| |
Collapse
|
40
|
Cauli E, Polidoro MA, Marzorati S, Bernardi C, Rasponi M, Lleo A. Cancer-on-chip: a 3D model for the study of the tumor microenvironment. J Biol Eng 2023; 17:53. [PMID: 37592292 PMCID: PMC10436436 DOI: 10.1186/s13036-023-00372-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
The approval of anticancer therapeutic strategies is still slowed down by the lack of models able to faithfully reproduce in vivo cancer physiology. On one hand, the conventional in vitro models fail to recapitulate the organ and tissue structures, the fluid flows, and the mechanical stimuli characterizing the human body compartments. On the other hand, in vivo animal models cannot reproduce the typical human tumor microenvironment, essential to study cancer behavior and progression. This study reviews the cancer-on-chips as one of the most promising tools to model and investigate the tumor microenvironment and metastasis. We also described how cancer-on-chip devices have been developed and implemented to study the most common primary cancers and their metastatic sites. Pros and cons of this technology are then discussed highlighting the future challenges to close the gap between the pre-clinical and clinical studies and accelerate the approval of new anticancer therapies in humans.
Collapse
Affiliation(s)
- Elisa Cauli
- Department of Electronics, Information and Bioengineering, Politecnico Di Milano, Milan, Italy.
- Accelera Srl, Nerviano, Milan, Italy.
| | - Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Simona Marzorati
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico Di Milano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
41
|
Loewa A, Feng JJ, Hedtrich S. Human disease models in drug development. NATURE REVIEWS BIOENGINEERING 2023; 1:1-15. [PMID: 37359774 PMCID: PMC10173243 DOI: 10.1038/s44222-023-00063-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 06/20/2023]
Abstract
Biomedical research is undergoing a paradigm shift towards approaches centred on human disease models owing to the notoriously high failure rates of the current drug development process. Major drivers for this transition are the limitations of animal models, which, despite remaining the gold standard in basic and preclinical research, suffer from interspecies differences and poor prediction of human physiological and pathological conditions. To bridge this translational gap, bioengineered human disease models with high clinical mimicry are being developed. In this Review, we discuss preclinical and clinical studies that benefited from these models, focusing on organoids, bioengineered tissue models and organs-on-chips. Furthermore, we provide a high-level design framework to facilitate clinical translation and accelerate drug development using bioengineered human disease models.
Collapse
Affiliation(s)
- Anna Loewa
- Department of Infectious Diseases and Respiratory Medicine, CharitĂ© â UniversitĂ€tsmedizin Berlin, corporate member of Freie UniversitĂ€t Berlin and Humboldt UniversitĂ€t zu Berlin, Berlin, Germany
| | - James J. Feng
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC Canada
- Department of Mathematics, University of British Columbia, Vancouver, BC Canada
| | - Sarah Hedtrich
- Department of Infectious Diseases and Respiratory Medicine, CharitĂ© â UniversitĂ€tsmedizin Berlin, corporate member of Freie UniversitĂ€t Berlin and Humboldt UniversitĂ€t zu Berlin, Berlin, Germany
- Center of Biological Design, Berlin Institute of Health at CharitĂ© â UniversitĂ€tsmedizin Berlin, corporate member of Freie UniversitĂ€t Berlin and Humboldt UniversitĂ€t zu Berlin, Berlin, Germany
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC Canada
- Max-DelbrĂŒck Center for Molecular Medicine (MCD), Helmholtz Association, Berlin, Germany
| |
Collapse
|
42
|
Sunildutt N, Parihar P, Chethikkattuveli Salih AR, Lee SH, Choi KH. Revolutionizing drug development: harnessing the potential of organ-on-chip technology for disease modeling and drug discovery. Front Pharmacol 2023; 14:1139229. [PMID: 37180709 PMCID: PMC10166826 DOI: 10.3389/fphar.2023.1139229] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
The inefficiency of existing animal models to precisely predict human pharmacological effects is the root reason for drug development failure. Microphysiological system/organ-on-a-chip technology (organ-on-a-chip platform) is a microfluidic device cultured with human living cells under specific organ shear stress which can faithfully replicate human organ-body level pathophysiology. This emerging organ-on-chip platform can be a remarkable alternative for animal models with a broad range of purposes in drug testing and precision medicine. Here, we review the parameters employed in using organ on chip platform as a plot mimic diseases, genetic disorders, drug toxicity effects in different organs, biomarker identification, and drug discoveries. Additionally, we address the current challenges of the organ-on-chip platform that should be overcome to be accepted by drug regulatory agencies and pharmaceutical industries. Moreover, we highlight the future direction of the organ-on-chip platform parameters for enhancing and accelerating drug discoveries and personalized medicine.
Collapse
Affiliation(s)
- Naina Sunildutt
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | - Pratibha Parihar
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | | | - Sang Ho Lee
- College of Pharmacy, Jeju National University, Jeju, Republic of Korea
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
43
|
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
|
44
|
CalĂ G, Sina B, De Coppi P, Giobbe GG, Gerli MFM. Primary human organoids models: Current progress and key milestones. Front Bioeng Biotechnol 2023; 11:1058970. [PMID: 36959902 PMCID: PMC10029057 DOI: 10.3389/fbioe.2023.1058970] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
During the past 10Â years the world has experienced enormous progress in the organoids field. Human organoids have shown huge potential to study organ development, homeostasis and to model diseases in vitro. The organoid technology has been widely and increasingly applied to generate patient-specific in vitro 3D cultures, starting from both primary and reprogrammed stem/progenitor cells. This has consequently fostered the development of innovative disease models and new regenerative therapies. Human primary, or adult stem/progenitor cell-derived, organoids can be derived from both healthy and pathological primary tissue samples spanning from fetal to adult age. The resulting 3D culture can be maintained for several months and even years, while retaining and resembling its original tissue's properties. As the potential of this technology expands, new approaches are emerging to further improve organoid applications in biology and medicine. This review discusses the main organs and tissues which, as of today, have been modelled in vitro using primary organoid culture systems. Moreover, we also discuss the advantages, limitations, and future perspectives of primary human organoids in the fields of developmental biology, disease modelling, drug testing and regenerative medicine.
Collapse
Affiliation(s)
- Giuseppe CalĂ
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, United Kingdom
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Beatrice Sina
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, United Kingdom
- Politecnico di Milano, Milano, Italy
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Giovanni Giuseppe Giobbe
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Mattia Francesco Maria Gerli
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, United Kingdom
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
45
|
Mohandas S, Gayatri V, Kumaran K, Gopinath V, Paulmurugan R, Ramkumar KM. New Frontiers in Three-Dimensional Culture Platforms to Improve Diabetes Research. Pharmaceutics 2023; 15:pharmaceutics15030725. [PMID: 36986591 PMCID: PMC10056755 DOI: 10.3390/pharmaceutics15030725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Diabetes mellitus is associated with defects in islet ÎČ-cell functioning and consequent hyperglycemia resulting in multi-organ damage. Physiologically relevant models that mimic human diabetic progression are urgently needed to identify new drug targets. Three-dimensional (3D) cell-culture systems are gaining a considerable interest in diabetic disease modelling and are being utilized as platforms for diabetic drug discovery and pancreatic tissue engineering. Three-dimensional models offer a marked advantage in obtaining physiologically relevant information and improve drug selectivity over conventional 2D (two-dimensional) cultures and rodent models. Indeed, recent evidence persuasively supports the adoption of appropriate 3D cell technology in ÎČ-cell cultivation. This review article provides a considerably updated view of the benefits of employing 3D models in the experimental workflow compared to conventional animal and 2D models. We compile the latest innovations in this field and discuss the various strategies used to generate 3D culture models in diabetic research. We also critically review the advantages and the limitations of each 3D technology, with particular attention to the maintenance of ÎČ-cell morphology, functionality, and intercellular crosstalk. Furthermore, we emphasize the scope of improvement needed in the 3D culture systems employed in diabetes research and the promises they hold as excellent research platforms in managing diabetes.
Collapse
Affiliation(s)
- Sundhar Mohandas
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vijaya Gayatri
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Kriya Kumaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vipin Gopinath
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Molecular Oncology Division, Malabar Cancer Centre, Moozhikkara P.O, Thalassery 670103, Kerala, India
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| |
Collapse
|
46
|
Amir S, Arathi A, Reshma S, Mohanan PV. Microfluidic devices for the detection of disease-specific proteins and other macromolecules, disease modelling and drug development: A review. Int J Biol Macromol 2023; 235:123784. [PMID: 36822284 DOI: 10.1016/j.ijbiomac.2023.123784] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023]
Abstract
Microfluidics is a revolutionary technology that has promising applications in the biomedical field.Integrating microfluidic technology with the traditional assays unravels the innumerable possibilities for translational biomedical research. Microfluidics has the potential to build up a novel platform for diagnosis and therapy through precise manipulation of fluids and enhanced throughput functions. The developments in microfluidics-based devices for diagnostics have evolved in the last decade and have been established for their rapid, effective, accurate and economic advantages. The efficiency and sensitivity of such devices to detect disease-specific macromolecules like proteins and nucleic acids have made crucial impacts in disease diagnosis. The disease modelling using microfluidic systems provides a more prominent replication of the in vivo microenvironment and can be a better alternative for the existing disease models. These models can replicate critical microphysiology like the dynamic microenvironment, cellular interactions, and biophysical and biochemical cues. Microfluidics also provides a promising system for high throughput drug screening and delivery applications. However, microfluidics-based diagnostics still encounter related challenges in the reliability, real-time monitoring and reproducibility that circumvents this technology from being impacted in the healthcare industry. This review highlights the recent microfluidics developments for modelling and diagnosing common diseases, including cancer, neurological, cardiovascular, respiratory and autoimmune disorders, and its applications in drug development.
Collapse
Affiliation(s)
- S Amir
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695 012, Kerala, India
| | - A Arathi
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695 012, Kerala, India
| | - S Reshma
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695 012, Kerala, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695 012, Kerala, India.
| |
Collapse
|
47
|
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]
|
48
|
Goldrick C, Guri I, Herrera-Oropeza G, OâBrien-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, Guyâs Campus, Kingâs College London, London, United Kingdom
| |
Collapse
|
49
|
Qiu L, Kong B, Kong T, Wang H. Recent advances in liver-on-chips: Design, fabrication, and applications. SMART MEDICINE 2023; 2:e20220010. [PMID: 39188562 PMCID: PMC11235950 DOI: 10.1002/smmd.20220010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/20/2022] [Indexed: 08/28/2024]
Abstract
The liver is a multifunctional organ and the metabolic center of the human body. Most drugs and toxins are metabolized in the liver, resulting in varying degrees of hepatotoxicity. The damage of liver will seriously affect human health, so it is very important to study the prevention and treatment of liver diseases. At present, there are many research studies in this field. However, most of them are based on animal models, which are limited by the time-consuming processes and species difference between human and animals. In recent years, liver-on-chips have emerged and developed rapidly and are expected to replace animal models. Liver-on-chips refer to the use of a small number of liver cells on the chips to simulate the liver microenvironment and ultrastructure in vivo. They hold extensive applications in multiple fields by reproducing the unique physiological functions of the liver in vitro. In this review, we first introduced the physiology and pathology of liver and then described the cell system of liver-on-chips, the chip-based liver models, and the applications of liver-on-chips in liver transplantation, drug screening, and metabolic evaluation. Finally, we discussed the currently encountered challenges and future trends in liver-on-chips.
Collapse
Affiliation(s)
- Linjie Qiu
- The Eighth Affiliated HospitalSun YatâSen UniversityShenzhenChina
- School of MedicineSun YatâSen UniversityShenzhenChina
| | - Bin Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingDepartment of Biomedical EngineeringSchool of MedicineShenzhen UniversityShenzhenChina
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingDepartment of Biomedical EngineeringSchool of MedicineShenzhen UniversityShenzhenChina
| | - Huan Wang
- The Eighth Affiliated HospitalSun YatâSen UniversityShenzhenChina
| |
Collapse
|
50
|
Tevlek A, Kecili S, Ozcelik OS, Kulah H, Tekin HC. Spheroid Engineering in Microfluidic Devices. ACS OMEGA 2023; 8:3630-3649. [PMID: 36743071 PMCID: PMC9893254 DOI: 10.1021/acsomega.2c06052] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/12/2022] [Indexed: 05/27/2023]
Abstract
Two-dimensional (2D) cell culture techniques are commonly employed to investigate biophysical and biochemical cellular responses. However, these culture methods, having monolayer cells, lack cell-cell and cell-extracellular matrix interactions, mimicking the cell microenvironment and multicellular organization. Three-dimensional (3D) cell culture methods enable equal transportation of nutrients, gas, and growth factors among cells and their microenvironment. Therefore, 3D cultures show similar cell proliferation, apoptosis, and differentiation properties to in vivo. A spheroid is defined as self-assembled 3D cell aggregates, and it closely mimics a cell microenvironment in vitro thanks to cell-cell/matrix interactions, which enables its use in several important applications in medical and clinical research. To fabricate a spheroid, conventional methods such as liquid overlay, hanging drop, and so forth are available. However, these labor-intensive methods result in low-throughput fabrication and uncontrollable spheroid sizes. On the other hand, microfluidic methods enable inexpensive and rapid fabrication of spheroids with high precision. Furthermore, fabricated spheroids can also be cultured in microfluidic devices for controllable cell perfusion, simulation of fluid shear effects, and mimicking of the microenvironment-like in vivo conditions. This review focuses on recent microfluidic spheroid fabrication techniques and also organ-on-a-chip applications of spheroids, which are used in different disease modeling and drug development studies.
Collapse
Affiliation(s)
- Atakan Tevlek
- METU
MEMS Research and Application Center, Ankara 06800, Turkey
| | - Seren Kecili
- The
Department of Bioengineering, Izmir Institute
of Technology, Urla, Izmir 35430, Turkey
| | - Ozge S. Ozcelik
- The
Department of Bioengineering, Izmir Institute
of Technology, Urla, Izmir 35430, Turkey
| | - Haluk Kulah
- METU
MEMS Research and Application Center, Ankara 06800, Turkey
- The
Department of Electrical and Electronics Engineering, Middle East Technical University, Ankara 06800, Turkey
| | - H. Cumhur Tekin
- METU
MEMS Research and Application Center, Ankara 06800, Turkey
- The
Department of Bioengineering, Izmir Institute
of Technology, Urla, Izmir 35430, Turkey
| |
Collapse
|