1
|
Ofori‐Kwafo A, Sigdel I, Al Mamun E, Zubcevic J, Tang Y. Gut-on-a-chip platforms: Bridging in vitro and in vivo models for advanced gastrointestinal research. Physiol Rep 2025; 13:e70356. [PMID: 40323242 PMCID: PMC12051376 DOI: 10.14814/phy2.70356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
The gastrointestinal (GI) tract plays a critical role in nutrient absorption, immune responses, and overall health. Traditional models such as two-dimensional cell cultures have provided valuable insights but fail to replicate the dynamic and complex microenvironment of the human gut. Gut-on-a-chip platforms, which incorporate cells located in the gut into microfluidic devices that simulate peristaltic motion and fluid flow, represent a significant advancement in modeling GI physiology and diseases. This review discusses the evolution of gut-on-a-chip technology, from simple cellular mono-cultures models to more sophisticated systems incorporating bi-cultures and tri-cultures that enable studies of drug metabolism, disease modeling, and gut-microbiome interactions. Although challenges remain, including maintaining long-term cell viability and replicating immune responses, these platforms hold great potential for advancing personalized medicine and improving drug discovery efforts targeting gastrointestinal disorders.
Collapse
Affiliation(s)
- Awurama Ofori‐Kwafo
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Indira Sigdel
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Earshed Al Mamun
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Jasenka Zubcevic
- University of South Florida Center for Microbiome ResearchMicrobiomes InstituteTampaFloridaUSA
- Department of Neurosurgery and Brain RepairUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Yuan Tang
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| |
Collapse
|
2
|
Schellberg BG, Koppes AN, Koppes RA. In situ monitoring of barrier function on-chip via automated, non-invasive luminescence sensing. LAB ON A CHIP 2025. [PMID: 40181784 PMCID: PMC11969330 DOI: 10.1039/d4lc01090f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Over the past 30 years, organs-on-a-chip (OOCs) have emerged as a robust alternative to address the technological challenges associated with current in vitro and in vivo options. Although OOCs offer improved bio-relevance and controlled complexity, broad adoption has remained limited. Most approaches to characterize on-chip structure and function require human intervention, limiting device translation and feasibility. Here, we introduce a new fiber optic-based sensing platform that enables automated, temporal luminescence sensing on-chip, validated for real-time readout of epithelial and endothelial barrier function under cytokine-induced inflammation. Our platform, capable of at least 1 μM resolution, tracked paracellular transport in situ for 9 days of culture under perfusion on-chip. These results offer an alternative sensing approach for continuous, non-invasive luminescence monitoring in OOCs.
Collapse
Affiliation(s)
- Bryan G Schellberg
- Department of Chemical Engineering, 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
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Mogas-Soldevila L, Zolotovsky K. Designing with Printed Responsive Biomaterials: A Review. 3D PRINTING AND ADDITIVE MANUFACTURING 2025; 12:155-168. [PMID: 40308656 PMCID: PMC12038318 DOI: 10.1089/3dp.2024.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
This review explores additive manufacturing (AM) strategies across disciplines for designing with responsive biomaterials and presents a vision of how printed responsive biomaterials (PRBs) can be integrated into everyday objects and buildings to enhance environmental and human health. Advancements in biomaterials science, biological materials manufacturing, synthetic biology, biomedical engineering, bio design, and living architecture are ushering in a new era characterized by multisensory interactions within everyday products and built environments. The material systems developed in recent research demonstrate the ability to interact with their environments through biological, chemical, or physical processes, yielding functionalities desirable in daily-use products. These include self-healing, health diagnostics, pathogen neutralization, adjustable stiffness, strain detection, threat visualization, shapeshifting, toxin trapping, stress correction, waste processing, and energy generation. Here we review examples of AM of biobased environmentally interactive materials using biopolymer composites, electrochemical and resistive devices, active molecules, bio sensors, living cells, spores, or cell-free sites, resulting in genetically active, and physical and chemical interactive systems. We highlight their robustness and evaluate their potential for scaling up into designs and architectures on Earth and beyond.
Collapse
Affiliation(s)
- Laia Mogas-Soldevila
- DumoLab Research, Department of Graduate Architecture, Stuart Weitzman School of Design, University of Pennsylvania, Philadelphia, PA, USA
| | - Katia Zolotovsky
- Department of Art and Design & Chemistry and Chemical Biology, Boston Campus, Northeastern University Boston, Massachusetts, USA
| |
Collapse
|
4
|
Boyanova L, Gergova R, Markovska R. Coculture systems to study interactions between anaerobic bacteria and intestinal epithelium. Anaerobe 2025; 92:102949. [PMID: 40010487 DOI: 10.1016/j.anaerobe.2025.102949] [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: 08/30/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Coculture systems (CCSs) are experimental tools used to study the interactions of anaerobic bacteria among themselves and the gut epithelial cells under conditions simulating the human gut, unlike those in animal models. Although the studies on animal models are useful in determining the relationship between the causative agents of infections and human infections, they have disadvantages, such as ethical issues, in addition to the differences in the microbiota of the animal and humans. Therefore, the results obtained using animal models cannot be directly extrapolated to humans. CCSs can more completely reflect in vivo gut homeostasis and contribute to better understanding of the interplay between the intestinal cells and anaerobes, prevalent among the gut bacteria. Moreover, they provide new insights on the pathogenesis of infections and aid in assessing the usefulness of new probiotics and antibacterials. Therefore, CCSs, including the gut-on-a-chip models, can significantly improve microbiota-based therapy. Moreover, they can also be used to detect microbiota-derived metabolites such as those with mutagenic properties. The aim of this review was to explore selected CCS models of anaerobes with intestinal epithelium and their application in investigating intestinal homeostasis. The focus was to highlight the application of different CCSs and important data obtained from their implementation.
Collapse
Affiliation(s)
- Lyudmila Boyanova
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria.
| | - Raina Gergova
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria
| | - Rumyana Markovska
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria
| |
Collapse
|
5
|
Moyer HL, Vergara L, Stephan C, Sakolish C, Ford LC, Tsai HHD, Lin HC, Chiu WA, Villenave R, Hewitt P, Ferguson SS, Rusyn I. Comparative analysis of Caco-2 cells and human jejunal and duodenal enteroid-derived cells in gel- and membrane-based barrier models of intestinal permeability. Toxicol Sci 2025; 204:181-197. [PMID: 39886939 PMCID: PMC11939079 DOI: 10.1093/toxsci/kfaf011] [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] [Indexed: 02/01/2025] Open
Abstract
Intestinal absorption is a key toxicokinetics parameter. Although the colon carcinoma cell line Caco-2 is the most used in vitro model to estimate human drug absorption, models representing other intestinal segments are available. We characterized the morphology, tissue-specific markers, and functionality of 3 human intestinal cell types: Caco-2, primary human enteroid-derived cells from jejunum (J2), and duodenum (D109) when cultured in the OrganoPlate 3-lane 40 microphysiological system (MPS) or static 24-well Transwells. In both conditions, J2 and D109 formed dome-like structures; Caco-2 formed uniform monolayers. In MPS, only Caco-2 formed tubules. Cells grown on Transwells formed a thicker monolayer. All cells and conditions exhibited expression of ZO-1 (tight junctions). Polarization markers Ezrin and Villin were highest in J2 and D109 in MPS, highest expression of Mucin was observed with J2. However, J2 and D109 exhibited poor barrier (70 kDa TRITC-dextran) in MPS, whereas robust barrier was recorded in Transwells. Barrier function and drug transport were evaluated using caffeine, indomethacin, and propranolol. The gel lane in MPS acted as a blockade; only a small fraction crossed, even without cells. The permeability ratios were used to parameterize the probabilistic compartmental absorption model to determine whether in vitro data could reduce uncertainty. The most accurate prediction of the fraction absorbed was achieved with Transwell-derived data from Caco-2, combined with the experimentally derived segment-specific absorption ratios. The impact of this study includes demonstration that enteroid-derived cells cultured in MPS show most physiological morphology, but that studies of drug permeability in this MPS are challenging.
Collapse
Affiliation(s)
- Haley L Moyer
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| | - Leoncio Vergara
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, United States
| | - Clifford Stephan
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, United States
| | - Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| | - Lucie C Ford
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| | - Han-Hsuan D Tsai
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| | - Hsing-Chieh Lin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| | - Weihsueh A Chiu
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel 4070, Switzerland
| | | | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States
| |
Collapse
|
6
|
Wang K, Wang Y, Han J, Liang Z, Zhang W, Li X, Chen J, Wang L. Biofabrication and simulation techniques for gut-on-a-chip. Biofabrication 2025; 17:022011. [PMID: 39965538 DOI: 10.1088/1758-5090/adb7c1] [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: 08/23/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Biomimetic gut models show promise for enhancing our understanding of intestinal disorder pathogenesis and accelerating therapeutic strategy development. Currentin vitromodels predominantly comprise traditional static cell culture and animal models. Static cell culture lacks the precise control of the complex microenvironment governing human intestinal function. Animal models provide greater microenvironment complexity but fail to accurately replicate human physiological conditions due to interspecies differences. As the available models do not accurately reflect the microphysiological environment and functions of the human intestine, their applications are limited. An optimal approach to intestinal modeling is yet to be developed, but the field will probably benefit from advances in biofabrication techniques. This review highlights biofabrication strategies for constructing biomimetic intestinal models and research approaches for simulating key intestinal physiological features. We also discuss potential biomedical applications of these models and provide an outlook on multi-scale intestinal modeling.
Collapse
Affiliation(s)
- Ke Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Zhixiang Liang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| |
Collapse
|
7
|
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
|
8
|
Han J, Wang Y, Ding J, Chen H, Shi C, Li X, Xu Z, Chen J, Kong F, Wang L. Gut-on-a-Chip Reveals Enhanced Peristalsis Reduces Nanoplastic-Induced Inflammation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408208. [PMID: 39587010 DOI: 10.1002/smll.202408208] [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: 09/10/2024] [Revised: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Nanoplastics (NPs) pollution is a global issue posing potential threats to human health, particularly the digestive system. NPs may exacerbate intestinal inflammation, increasing the risk of inflammatory bowel disease. However, the impact of intestinal peristalsis on NP-induced inflammation remains unknown. Here, a biomimetic gut-on-a-chip (GOC) with integrated online sensing is presented to investigate NPs' impact on intestinal inflammation and propose enhanced peristalsis as a potential intervention. The GOC simulates intestinal peristalsis through periodic stretching and the optimized sensors dynamically detect inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) over 12 d with low detection limits (0.0095 and 0.0020 pg mL-1). Exposure to NPs led to vacuolization, apoptosis, and loss of tight junction proteins in intestinal cells, with IL-6 and TNF-α secretion peaking at 24 h (1341.55 ± 64.91 and 862.03 ± 66.45 pg mL-1). Notably, increasing periodic strain alleviates inflammatory cytokines secretion induced by NPs. With strain increased from 5% to 6.5%, IL-6 and TNF-α secretion decrease by 2.73-fold and 3.34-fold, respectively. This highlights the protective role of intestinal peristalsis in reducing NP-induced inflammation.
Collapse
Affiliation(s)
- Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250353, China
- The Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, School of Mechanical Engineering, Tianjin University, Tianjin, 300072, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250353, China
| | - Jiemeng Ding
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250353, China
| | - Hao Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250353, China
| | - Chaoyang Shi
- The Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, School of Mechanical Engineering, Tianjin University, Tianjin, 300072, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhipeng Xu
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Medical School, Sheffield, S10 2RX, UK
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250353, China
| | - Feng Kong
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250353, China
| |
Collapse
|
9
|
Moro LG, Guarnier LP, Azevedo MF, Fracasso JAR, Lucio MA, de Castro MV, Dias ML, Lívero FADR, Ribeiro-Paes JT. A Brief History of Cell Culture: From Harrison to Organs-on-a-Chip. Cells 2024; 13:2068. [PMID: 39768159 PMCID: PMC11674496 DOI: 10.3390/cells13242068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 01/11/2025] Open
Abstract
This comprehensive overview of the historical milestones in cell culture underscores key breakthroughs that have shaped the field over time. It begins with Wilhelm Roux's seminal experiments in the 1880s, followed by the pioneering efforts of Ross Granville Harrison, who initiated groundbreaking experiments that fundamentally shaped the landscape of cell culture in the early 20th century. Carrel's influential contributions, notably the immortalization of chicken heart cells, have marked a significant advancement in cell culture techniques. Subsequently, Johannes Holtfreter, Aron Moscona, and Joseph Leighton introduced methodological innovations in three-dimensional (3D) cell culture, initiated by Alexis Carrel, laying the groundwork for future consolidation and expansion of the use of 3D cell culture in different areas of biomedical sciences. The advent of induced pluripotent stem cells by Takahashi and Yamanaka in 2006 was revolutionary, enabling the reprogramming of differentiated cells into a pluripotent state. Since then, recent innovations have included spheroids, organoids, and organ-on-a-chip technologies, aiming to mimic the structure and function of tissues and organs in vitro, pushing the boundaries of biological modeling and disease understanding. In this review, we overview the history of cell culture shedding light on the main discoveries, pitfalls and hurdles that were overcome during the transition from 2D to 3D cell culture techniques. Finally, we discussed the future directions for cell culture research that may accelerate the development of more effective and personalized treatments.
Collapse
Affiliation(s)
- Lincoln Gozzi Moro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Lucas Pires Guarnier
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
| | | | | | - Marco Aurélio Lucio
- Graduate Program in Environment and Regional Development, University of Western São Paulo, Presidente Prudente 19050-920, Brazil;
| | - Mateus Vidigal de Castro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Marlon Lemos Dias
- Precision Medicine Research Center, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro—UFRJ, Rio de Janeiro 21941-630, Brazil;
| | | | - João Tadeu Ribeiro-Paes
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
- Laboratory of Genetics and Cell Therapy (GenTe Cel), Department of Biotechnology, São Paulo State University—UNESP, Assis 19806-900, Brazil
| |
Collapse
|
10
|
Jang Y, Kim H, Oh J. An Array of Carbon Nanofiber Bundle_Based 3D In Vitro Intestinal Microvilli for Mimicking Functional and Physical Activities of the Small Intestine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404842. [PMID: 39212639 DOI: 10.1002/smll.202404842] [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: 06/13/2024] [Revised: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Researchers have developed in vitro small intestine models of biomimicking microvilli, such as gut-on-a-chip devices. However, fabrication methods developed to date for 2D and 3D in vitro gut still have unsolved limitations. In this study, an innovative fabrication method of a 3D in vitro gut model is introduced for effective drug screening. The villus is formed on a patterned carbon nanofiber (CNF) bundle as a flexible and biocompatible scaffold. Mechanical properties of the fabricated villi structure are investigates. A microfluidic system is applied to induce the movement of CNFs villi. F-actin and Occludin staining of Caco-2 cells on a 2D flat-chip as a control and a 3D gut-chip with or without fluidic stress is observed. A permeability test of FD20 is performed. The proposed 3D gut-chip with fluidic stress achieve the highest value of Papp. Mechano-active stimuli caused by distinct structural and movement effects of CNFs villi as well as stiffness of the suggested CNFs villi not only can help accelerate cell differentiation but also can improve permeability. The proposed 3D gut-chip system further strengthens the potential of the platform to increase the accuracy of various drug tests.
Collapse
Affiliation(s)
- Yeongseok Jang
- Department of Mechanical Design Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Hyojae Kim
- Center for Social Innovation Policy, Office of S&T Policy Planning, Korea Institute of S&T Evaluation and Planning, Eumseong, 27740, Republic of Korea
| | - Jonghyun Oh
- Department of Nano-Bio Mechanical System Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| |
Collapse
|
11
|
Wang X, Zhu Y, Cheng Z, Zhang C, Liao Y, Liu B, Zhang D, Li Z, Fang Y. Emerging microfluidic gut-on-a-chip systems for drug development. Acta Biomater 2024; 188:48-64. [PMID: 39299625 DOI: 10.1016/j.actbio.2024.09.012] [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: 05/16/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
The gut is a vital organ that is central to the absorption and metabolic processing of orally administered drugs. While there have been many models developed with the goal of studying the absorption of drugs in the gut, these models fail to adequately recapitulate the diverse, complex gastrointestinal microenvironment. The recent emergence of microfluidic organ-on-a-chip technologies has provided a novel means of modeling the gut, yielding radical new insights into the structure of the gut and the mechanisms through which it shapes disease, with key implications for biomedical developmental efforts. Such organ-on-a-chip technologies have been demonstrated to exhibit greater cost-effectiveness, fewer ethical concerns, and a better ability to address inter-species differences in traditional animal models in the context of drug development. The present review offers an overview of recent developments in the reconstruction of gut structure and function in vitro using microfluidic gut-on-a-chip (GOC) systems, together with a discussion of the potential applications of these platforms in the context of drug development and the challenges and future prospects associated with this technology. STATEMENT OF SIGNIFICANCE: This paper outlines the characteristics of the different cell types most frequently used to construct microfluidic gut-on-a-chip models and the microfluidic devices employed for the study of drug absorption. And the applications of gut-related multichip coupling and disease modelling in the context of drug development is systematically reviewed. With the detailed summarization of microfluidic chip-based gut models and discussion of the prospective directions for practical application, this review will provide insights to the innovative design and application of microfluidic gut-on-a-chip for drug development.
Collapse
Affiliation(s)
- Xueqi Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Yuzhuo Zhu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Zhaoming Cheng
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Chuanjun Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Yumeng Liao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Boshi Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Yuxin Fang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China; Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
12
|
Man Y, Liu Y, Chen Q, Zhang Z, Li M, Xu L, Tan Y, Liu Z. Organoids-On-a-Chip for Personalized Precision Medicine. Adv Healthc Mater 2024:e2401843. [PMID: 39397335 DOI: 10.1002/adhm.202401843] [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: 06/01/2024] [Revised: 08/25/2024] [Indexed: 10/15/2024]
Abstract
The development of personalized precision medicine has become a pivotal focus in modern healthcare. Organoids-on-a-Chip (OoCs), a groundbreaking fusion of organoid culture and microfluidic chip technology, has emerged as a promising approach to advancing patient-specific treatment strategies. In this review, the diverse applications of OoCs are explored, particularly their pivotal role in personalized precision medicine, and their potential as a cutting-edge technology is highlighted. By utilizing patient-derived organoids, OoCs offer a pathway to optimize treatments, create precise disease models, investigate disease mechanisms, conduct drug screenings, and individualize therapeutic strategies. The emphasis is on the significance of this technological fusion in revolutionizing healthcare and improving patient outcomes. Furthermore, the transformative potential of personalized precision medicine, future prospects, and ongoing advancements in the field, with a focus on genomic medicine, multi-omics integration, and ethical frameworks are discussed. The convergence of these innovations can empower patients, redefine treatment approaches, and shape the future of healthcare.
Collapse
Affiliation(s)
- Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
13
|
Ebrahimi A, Ghorbanpoor H, Apaydın E, Demir Cevizlidere B, Özel C, Tüfekçioğlu E, Koç Y, Topal AE, Tomsuk Ö, Güleç K, Abdullayeva N, Kaya M, Ghorbani A, Şengel T, Benzait Z, Uysal O, Eker Sarıboyacı A, Doğan Güzel F, Singh H, Hassan S, Ankara H, Pat S, Atalay E, Avci H. Convenient rapid prototyping microphysiological niche for mimicking liver native basement membrane: Liver sinusoid on a chip. Colloids Surf B Biointerfaces 2024; 245:114292. [PMID: 39383580 DOI: 10.1016/j.colsurfb.2024.114292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Liver is responsible for the metabolization processes of up to 90 % of compounds and toxins in the body. Therefore liver-on-a-chip systems, as an in vitro promising cell culture platform, have great importance for fundamental science and drug development. In most of the liver-on-a-chip studies, seeding cells on both sides of a porous membrane, which represents the basement membrane, fail to resemble the native characteristics of biochemical, biophysical, and mechanical properties. In this study, polycarbonate (PC) and polyethylene terephthalate (PET) membranes were coated with gelatin to address this issue by accurately mimicking the native basement membrane present in the space of Disse. Various coating methods were used, including doctor blade, gel micro-injection, electrospinning, and spin coating. Spin coating was demonstrated to be the most effective technique owing to the ability to produce thin gel thickness with desirable surface roughness for cell interactions on both sides of the membrane. HepG2 and EA.HY926 cells were seeded on the upper and bottom sides of the gelatin-coated PET membrane and cultured on-chip for 7 days. Cell viability increased from 90 % to 95 %, while apoptotic index decreased. Albumin secretion notably rose between days 1-7 and 4-7, while GST-α secretion decreased from day 1 to day 7. In conclusion, the optimized spin coating process reported here can effectively modify the membranes to better mimic the native basement membrane niche characteristics.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Hamed Ghorbanpoor
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biomedical Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Elif Apaydın
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Bahar Demir Cevizlidere
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ceren Özel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Emre Tüfekçioğlu
- Department of Industrial Design/Department of Industrial Design, Faculty of Architecture and Design, Eskisehir Technical University, Eskisehir, Türkiye
| | - Yücel Koç
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ahmet Emin Topal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, School of Pharmacy, Bahçeşehir University, Istanbul, Türkiye
| | - Özlem Tomsuk
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Mechanical Engineering, Middle East Technical University, Ankara 06800, Türkiye
| | - Kadri Güleç
- Department of Analytical Chemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Nuran Abdullayeva
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Murat Kaya
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Aynaz Ghorbani
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Tayfun Şengel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Central Research Laboratory Research and Application Center (ARUM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Zineb Benzait
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ayla Eker Sarıboyacı
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Fatma Doğan Güzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, Ankara, Türkiye
| | - Hemant Singh
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Hüseyin Ankara
- Mining Engineering Department, Engineering-Architecture Faculty, Eskisehir Osmangazi University, Meşelik Campus, Eskisehir 26480, Türkiye
| | - Suat Pat
- Eskisehir Osmangazi University, Faculty of Science, Department of Physics, Eskisehir TR-26040, Türkiye
| | - Eray Atalay
- Department of Ophthalmology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir 26040, Türkiye
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye; Translational Medicine Research and Clinical Center (TATUM), Eskisehir Osmangazi University, Eskisehir, Türkiye.
| |
Collapse
|
14
|
Bucking C, Bury NR, Sundh H, Wood CM. Making in vitro conditions more reflective of in vivo conditions for research on the teleost gastrointestinal tract. J Exp Biol 2024; 227:jeb246440. [PMID: 39392112 PMCID: PMC11529878 DOI: 10.1242/jeb.246440] [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] [Indexed: 10/12/2024]
Abstract
To date, the majority of in vitro or ex vivo fish gastrointestinal research has been conducted under unrealistic conditions. In a living fish, ionic conditions, as well as levels of ammonia, pH, HCO3- and PCO2 differ considerably between the different regions of the gastrointestinal tract. These factors also differ from those of the saline often used in gut research. Furthermore, the oxygen gradient from the serosa to the gut lumen is rarely considered: in contrast to the serosa, the lumen is a hypoxic/anoxic environment. In addition, the gut microbiome plays a significant role in gut physiology, increasing the complexity of the in vivo gut, but replicating the microbial community for in vitro studies is exceptionally difficult. However, there are ways in which we can begin to overcome these challenges. Firstly, the luminal chemistry and PO2 in each gut compartment must be carefully considered. Secondly, although microbiological culture techniques are improving, we must learn how to maintain the microbiome diversity seen in vivo. Finally, for ex vivo studies, developing mucosal (luminal) solutions that more closely mimic the in vivo conditions will better replicate physiological processes. Within the field of mammalian gut physiology, great advances in 'gut-on-chip' devices are providing the tools to better replicate in vivo conditions; adopting and adapting this technology may assist in fish gut research initiatives. This Commentary aims to make fish gut physiologists aware of the various issues in replicating the in vivo conditions and identifies solutions as well as those areas that require further improvement.
Collapse
Affiliation(s)
- Carol Bucking
- Department of Biology, Farquharson Life Science Building, York University, Toronto, ON, M3J 1P3, Canada
| | - Nic R. Bury
- School of Ocean and Earth Sciences, University of Southampton, National Oceanographic Centre, Waterfront Campus, Southampton, Hampshire, SO14 3ZH, UK
| | - Henrik Sundh
- Department of Biological & Environmental Sciences, University of Gothenburg, Medicinaregatan 7 B, 41390 Göteborg, Sweden
| | - Chris M. Wood
- Department of Zoology, University of British Columbia, 6270 University Blvd, Vancouver, BC, V6T1Z4, Canada
| |
Collapse
|
15
|
Pan X, Chen J, Han J, Zhang W, Su W, Xu Z, Li X, Song M, Song W, Xie X, Wang L. Critical Suitability Evaluation of Caco-2 Cells for Gut-on-a-Chip. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51139-51149. [PMID: 39265077 DOI: 10.1021/acsami.4c11409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Currently, culturing Caco-2 cells in a Gut-on-a-chip (GOC) is well-accepted for developing intestinal disease models and drug screening. However, Caco-2 cells were found to overexpress surface proteins (e.g., P-gp) compared with the normal intestinal epithelial cells in vivo. To critically evaluate the challenge and suitability of Caco-2 cells, a GOC integrated with a carcinoembryonic antigen (CEA) biosensor was developed. This three-electrode system electrochemical sensor detects CEA by antigen-antibody specific binding, and it exhibits high selectivity, excellent stability, and good reproducibility. Under dynamic culturing in the GOC, Caco-2 cells exhibited an intestinal villus-like structure and maintained tissue barrier integrity. Meanwhile, CEA was discovered to be secreted from 0 to 0.22 ng/mL during the 10-day culturing of Caco-2 cells. Especially, CEA secretion increased significantly with the differentiation of Caco-2 cells after 6 days of culturing. The sustained high-level CEA secretion may induce cells to avoid apoptotic stimuli, which faithfully reflects the efficacy of a new drug and the mechanism of intestinal disease. Different kinds of cell types (e.g., intestinal primary cells, stem cell-induced differentiation) in the GOC should be attempted for drug screening in the future.
Collapse
Affiliation(s)
- Xiatong Pan
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenxian Zhang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Weiguang Su
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Zhipeng Xu
- School of Medicine & Population Health, University of Sheffield, Sheffield S10 2RX, U.K
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Ming Song
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wei Song
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
16
|
Teo AJT, Ng SK, Khoo K, Wong SH, Li KHH. Microfluidic Gastrointestinal Cell Culture Technologies-Improvements in the Past Decade. BIOSENSORS 2024; 14:449. [PMID: 39329824 PMCID: PMC11429516 DOI: 10.3390/bios14090449] [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: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Gastrointestinal cell culture technology has evolved in the past decade with the integration of microfluidic technologies, bringing advantages with greater selectivity and cost effectiveness. Herein, these technologies are sorted into three categories, namely the cell-culture insert devices, conventional microfluidic devices, and 3D-printed microfluidic devices. Each category is discussed in brief with improvements also discussed here. Introduction of different companies and applications derived from each are also provided to encourage uptake. Subsequently, future perspectives of integrating microfluidics with trending topics like stool-derived in vitro communities and gut-immune-tumor axis investigations are discussed. Insights on modular microfluidics and its implications on gastrointestinal cell cultures are also discussed here. Future perspectives on point-of-care (POC) applications in relations to gastrointestinal microfluidic devices are also discussed here. In conclusion, this review presents an introduction of each microfluidic platform with an insight into the greater contribution of microfluidics in gastrointestinal cell cultures.
Collapse
Affiliation(s)
- Adrian J. T. Teo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore; (A.J.T.T.); (K.K.)
| | - Siu-Kin Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (S.-K.N.); (S.H.W.)
| | - Kaydeson Khoo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore; (A.J.T.T.); (K.K.)
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (S.-K.N.); (S.H.W.)
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore; (A.J.T.T.); (K.K.)
| |
Collapse
|
17
|
Žukauskaitė K, Li M, Horvath A, Jarmalaitė S, Stadlbauer V. Cellular and Microbial In Vitro Modelling of Gastrointestinal Cancer. Cancers (Basel) 2024; 16:3113. [PMID: 39272971 PMCID: PMC11394127 DOI: 10.3390/cancers16173113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Human diseases are multifaceted, starting with alterations at the cellular level, damaging organs and their functions, and disturbing interactions and immune responses. In vitro systems offer clarity and standardisation, which are crucial for effectively modelling disease. These models aim not to replicate every disease aspect but to dissect specific ones with precision. Controlled environments allow researchers to isolate key variables, eliminate confounding factors and elucidate disease mechanisms more clearly. Technological progress has rapidly advanced model systems. Initially, 2D cell culture models explored fundamental cell interactions. The transition to 3D cell cultures and organoids enabled more life-like tissue architecture and enhanced intercellular interactions. Advanced bioreactor-based devices now recreate the physicochemical environments of specific organs, simulating features like perfusion and the gastrointestinal tract's mucus layer, enhancing physiological relevance. These systems have been simplified and adapted for high-throughput research, marking significant progress. This review focuses on in vitro systems for modelling gastrointestinal tract cancer and the side effects of cancer treatment. While cell cultures and in vivo models are invaluable, our main emphasis is on bioreactor-based in vitro modelling systems that include the gut microbiome.
Collapse
Affiliation(s)
- Kristina Žukauskaitė
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
| | - Melissa Li
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Biotech Campus Tulln, Fachhochschule Wiener Neustadt, 3430 Tulln, Austria
| | - Angela Horvath
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Center for Biomarker Research in Medicine (CBmed GmbH), 8010 Graz, Austria
| | - Sonata Jarmalaitė
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
- National Cancer Institute, 08406 Vilnius, Lithuania
| | - Vanessa Stadlbauer
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Center for Biomarker Research in Medicine (CBmed GmbH), 8010 Graz, Austria
| |
Collapse
|
18
|
Yuan S, Yuan H, Hay DC, Hu H, Wang C. Revolutionizing Drug Discovery: The Impact of Distinct Designs and Biosensor Integration in Microfluidics-Based Organ-on-a-Chip Technology. BIOSENSORS 2024; 14:425. [PMID: 39329800 PMCID: PMC11430660 DOI: 10.3390/bios14090425] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Traditional drug development is a long and expensive process with high rates of failure. This has prompted the pharmaceutical industry to seek more efficient drug development frameworks, driving the emergence of organ-on-a-chip (OOC) based on microfluidic technologies. Unlike traditional animal experiments, OOC systems provide a more accurate simulation of human organ microenvironments and physiological responses, therefore offering a cost-effective and efficient platform for biomedical research, particularly in the development of new medicines. Additionally, OOC systems enable quick and real-time analysis, high-throughput experimentation, and automation. These advantages have shown significant promise in enhancing the drug development process. The success of an OOC system hinges on the integration of specific designs, manufacturing techniques, and biosensors to meet the need for integrated multiparameter datasets. This review focuses on the manufacturing, design, sensing systems, and applications of OOC systems, highlighting their design and sensing capabilities, as well as the technical challenges they currently face.
Collapse
Affiliation(s)
- Sheng Yuan
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Zhejiang University, Haining 314400, China
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| |
Collapse
|
19
|
Shin YC, Than N, Park SJ, Kim HJ. Bioengineered human gut-on-a-chip for advancing non-clinical pharmaco-toxicology. Expert Opin Drug Metab Toxicol 2024; 20:593-606. [PMID: 38849312 DOI: 10.1080/17425255.2024.2365254] [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: 02/09/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION There is a growing need for alternative models to advance current non-clinical experimental models because they often fail to accurately predict drug responses in human clinical trials. Human organ-on-a-chip models have emerged as promising approaches for advancing the predictability of drug behaviors and responses. AREAS COVERED We summarize up-to-date human gut-on-a-chip models designed to demonstrate intricate interactions involving the host, microbiome, and pharmaceutical compounds since these models have been reported a decade ago. This overview covers recent advances in gut-on-a-chip models as a bridge technology between non-clinical and clinical assessments of drug toxicity and metabolism. We highlight the promising potential of gut-on-a-chip platforms, offering a reliable and valid framework for investigating reciprocal crosstalk between the host, gut microbiome, and drug compounds. EXPERT OPINION Gut-on-a-chip platforms can attract multiple end users as predictive, human-relevant, and non-clinical model. Notably, gut-on-a-chip platforms provide a unique opportunity to recreate a human intestinal microenvironment, including dynamic bowel movement, luminal flow, oxygen gradient, host-microbiome interactions, and disease-specific manipulations restricted in animal and in vitro cell culture models. Additionally, given the profound impact of the gut microbiome on pharmacological bioprocess, it is critical to leverage breakthroughs of gut-on-a-chip technology to address knowledge gaps and drive innovations in predictive drug toxicology and metabolism.
Collapse
Affiliation(s)
- Yong Cheol Shin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nam Than
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Soo Jin Park
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hyun Jung Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Inflammation and Immunity, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
20
|
Wang H, Li X, Shi P, You X, Zhao G. Establishment and evaluation of on-chip intestinal barrier biosystems based on microfluidic techniques. Mater Today Bio 2024; 26:101079. [PMID: 38774450 PMCID: PMC11107260 DOI: 10.1016/j.mtbio.2024.101079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
As a booming engineering technology, the microfluidic chip has been widely applied for replicating the complexity of human intestinal micro-physiological ecosystems in vitro. Biosensors, 3D imaging, and multi-omics have been applied to engineer more sophisticated intestinal barrier-on-chip platforms, allowing the improved monitoring of physiological processes and enhancing chip performance. In this review, we report cutting-edge advances in the microfluidic techniques applied for the establishment and evaluation of intestinal barrier platforms. We discuss different design principles and microfabrication strategies for the establishment of microfluidic gut barrier models in vitro. Further, we comprehensively cover the complex cell types (e.g., epithelium, intestinal organoids, endothelium, microbes, and immune cells) and controllable extracellular microenvironment parameters (e.g., oxygen gradient, peristalsis, bioflow, and gut-organ axis) used to recapitulate the main structural and functional complexity of gut barriers. We also present the current multidisciplinary technologies and indicators used for evaluating the morphological structure and barrier integrity of established gut barrier models in vitro. Finally, we highlight the challenges and future perspectives for accelerating the broader applications of these platforms in disease simulation, drug development, and personalized medicine. Hence, this review provides a comprehensive guide for the development and evaluation of microfluidic-based gut barrier platforms.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Pengcheng Shi
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- CAS-Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
21
|
Wang H, Kim R, Wang Y, Furtado KL, Sims CE, Tamayo R, Allbritton NL. In vitro co-culture of Clostridium scindens with primary human colonic epithelium protects the epithelium against Staphylococcus aureus. Front Bioeng Biotechnol 2024; 12:1382389. [PMID: 38681959 PMCID: PMC11045926 DOI: 10.3389/fbioe.2024.1382389] [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: 02/05/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
A complex and dynamic network of interactions exists between human gastrointestinal epithelium and intestinal microbiota. Therefore, comprehending intestinal microbe-epithelial cell interactions is critical for the understanding and treatment of intestinal diseases. Primary human colonic epithelial cells derived from a healthy human donor were co-cultured with Clostridium scindens (C. scindens), a probiotic obligate anaerobe; Staphylococcus aureus (S. aureus), a facultative anaerobe and intestinal pathogen; or both bacterial species in tandem. The co-culture hanging basket platform used for these experiments possessed walls of controlled oxygen (O2) permeability to support the formation of an O2 gradient across the intestinal epithelium using cellular O2 consumption, resulting in an anaerobic luminal and aerobic basal compartment. Both the colonic epithelial cells and C. scindens remained viable over 48 h during co-culture. In contrast, co-culture with S. aureus elicited significant damage to colonic epithelial cells within 24 h. To explore the influence of the intestinal pathogen on the epithelium in the presence of the probiotic bacteria, colonic epithelial cells were inoculated sequentially with the two bacterial species. Under these conditions, C. scindens was capable of repressing the production of S. aureus enterotoxin. Surprisingly, although C. scindens converted cholic acid to secondary bile acids in the luminal medium, the growth of S. aureus was not significantly inhibited. Nevertheless, this combination of probiotic and pathogenic bacteria was found to benefit the survival of the colonic epithelial cells compared with co-culture of the epithelial cells with S. aureus alone. This platform thus provides an easy-to-use and low-cost tool to study the interaction between intestinal bacteria and colonic cells in vitro to better understand the interplay of intestinal microbiota with human colonic epithelium.
Collapse
Affiliation(s)
- Hao Wang
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Raehyun Kim
- Department of Bioengineering, University of Washington, Seattle, WA, United States
- Department of Biological and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Kathleen L. Furtado
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Christopher E. Sims
- Department of Bioengineering, University of Washington, Seattle, WA, United States
- Department of Medicine/Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Rita Tamayo
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Nancy L. Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
22
|
Vera D, García-Díaz M, Torras N, Castillo Ó, Illa X, Villa R, Alvarez M, Martinez E. A 3D bioprinted hydrogel gut-on-chip with integrated electrodes for transepithelial electrical resistance (TEER) measurements. Biofabrication 2024; 16:035008. [PMID: 38574551 DOI: 10.1088/1758-5090/ad3aa4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/04/2024] [Indexed: 04/06/2024]
Abstract
Conventional gut-on-chip (GOC) models typically represent the epithelial layer of the gut tissue, neglecting other important components such as the stromal compartment and the extracellular matrix (ECM) that play crucial roles in maintaining intestinal barrier integrity and function. These models often employ hard, flat porous membranes for cell culture, thus failing to recapitulate the soft environment and complex 3D architecture of the intestinal mucosa. Alternatively, hydrogels have been recently introduced in GOCs as ECM analogs to support the co-culture of intestinal cells inin vivo-like configurations, and thus opening new opportunities in the organ-on-chip field. In this work, we present an innovative GOC device that includes a 3D bioprinted hydrogel channel replicating the intestinal villi architecture containing both the epithelial and stromal compartments of the gut mucosa. The bioprinted hydrogels successfully support both the encapsulation of fibroblasts and their co-culture with intestinal epithelial cells under physiological flow conditions. Moreover, we successfully integrated electrodes into the microfluidic system to monitor the barrier formation in real time via transepithelial electrical resistance measurements.
Collapse
Affiliation(s)
- Daniel Vera
- Instituto de Microelectrónica de Barcelona, IMB-CNM (CSIC), Bellaterra, Barcelona 08193, Spain
| | - María García-Díaz
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Núria Torras
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Óscar Castillo
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Xavi Illa
- Instituto de Microelectrónica de Barcelona, IMB-CNM (CSIC), Bellaterra, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08193, Spain
| | - Rosa Villa
- Instituto de Microelectrónica de Barcelona, IMB-CNM (CSIC), Bellaterra, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08193, Spain
| | - Mar Alvarez
- Instituto de Microelectrónica de Barcelona, IMB-CNM (CSIC), Bellaterra, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08193, Spain
| | - Elena Martinez
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08193, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona 08028, Spain
| |
Collapse
|
23
|
Mahieu L, Van Moll L, De Vooght L, Delputte P, Cos P. In vitro modelling of bacterial pneumonia: a comparative analysis of widely applied complex cell culture models. FEMS Microbiol Rev 2024; 48:fuae007. [PMID: 38409952 PMCID: PMC10913945 DOI: 10.1093/femsre/fuae007] [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: 10/02/2023] [Revised: 01/29/2024] [Accepted: 02/24/2024] [Indexed: 02/28/2024] Open
Abstract
Bacterial pneumonia greatly contributes to the disease burden and mortality of lower respiratory tract infections among all age groups and risk profiles. Therefore, laboratory modelling of bacterial pneumonia remains important for elucidating the complex host-pathogen interactions and to determine drug efficacy and toxicity. In vitro cell culture enables for the creation of high-throughput, specific disease models in a tightly controlled environment. Advanced human cell culture models specifically, can bridge the research gap between the classical two-dimensional cell models and animal models. This review provides an overview of the current status of the development of complex cellular in vitro models to study bacterial pneumonia infections, with a focus on air-liquid interface models, spheroid, organoid, and lung-on-a-chip models. For the wide scale, comparative literature search, we selected six clinically highly relevant bacteria (Pseudomonas aeruginosa, Mycoplasma pneumoniae, Haemophilus influenzae, Mycobacterium tuberculosis, Streptococcus pneumoniae, and Staphylococcus aureus). We reviewed the cell lines that are commonly used, as well as trends and discrepancies in the methodology, ranging from cell infection parameters to assay read-outs. We also highlighted the importance of model validation and data transparency in guiding the research field towards more complex infection models.
Collapse
Affiliation(s)
- Laure Mahieu
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Laurence Van Moll
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Linda De Vooght
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Paul Cos
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
24
|
Vorländer D, Schultz G, Hoffmann K, Rasch D, Dohnt K. PETR: A novel peristaltic mixed tubular bioreactor simulating human colonic conditions. Biotechnol Bioeng 2024; 121:1118-1143. [PMID: 38151924 DOI: 10.1002/bit.28636] [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: 07/07/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/29/2023]
Abstract
A novel bioreactor simulating human colonic conditions for in vitro cultivation of intestinal microbiota is presented. The PEristaltic mixed Tubular bioReactor (PETR) is modular designed and periodically kneaded to simulate intestinal peristalsis. The reactor is introduced, characterized from a bioprocess engineer's perspective and discussed in its ability to mimic colon conditions. PETR provides physiological temperature and appropriate anaerobic conditions, simulates intestinal peristalsis, and has a mean residence time of 32.8 ± 0.8 h comparable to the adult human colon. The single-tube design enables a time-constant and longitudinally progressive pH gradient from 5.5 to 7.0. Using a dialysis liquid containing high molecular weight polyethylene glycol, the integrated dialysis system efficiently absorbs short chain fatty acids (up to 60%) and water (on average 850 mL d-1 ). Cultivation of a typical gut bacterium (Bifidobacterium animalis) was performed to demonstrate the applicability for controlled microbiota cultivation. PETR is unique in combining simulation of the entire colon, peristaltic mixing, dialytic water and metabolite absorption, and a progressive pH gradient in a single-tube design. PETR is a further step to precise replication of colonic conditions in vitro for reliable and reproducible microbiota research, such as studying the effect of food compounds, prebiotics or probiotics, or the development and treatment of infections with enteric pathogens, but also for further medical applications such as drug delivery studies or to study the effect of drugs on and their degradation by the microbiota.
Collapse
Affiliation(s)
- David Vorländer
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Gábor Schultz
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Kristin Hoffmann
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Detlev Rasch
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Katrin Dohnt
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
25
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
26
|
González A, Fullaondo A, Odriozola A. Techniques, procedures, and applications in microbiome analysis. ADVANCES IN GENETICS 2024; 111:81-115. [PMID: 38908906 DOI: 10.1016/bs.adgen.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Microbiota is a complex community of microorganisms living in a defined environment. Until the 20th century, knowledge of microbiota was partial, as the techniques available for their characterization were primarily based on bacteriological culture. In the last twenty years, the development of DNA sequencing technologies, multi-omics, and bioinformatics has expanded our understanding of microorganisms. We have moved from mainly considering them isolated disease-causing agents to recognizing the microbiota as an essential component of host biology. These techniques have shown that the microbiome plays essential roles in various host phenotypes, influencing development, physiology, reproduction, and evolution. This chapter provides researchers with a summary of the primary concepts, sample collection, experimental techniques, and bioinformatics analysis commonly used in microbiome research. The main features, applications in microbiome studies, and their advantages and limitations are included in each section.
Collapse
Affiliation(s)
- Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Asier Fullaondo
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
27
|
Guo Y, Xie Y, Qin J. A generic pump-free organ-on-a-chip platform for assessment of intestinal drug absorption. Biotechnol J 2024; 19:e2300390. [PMID: 38375564 DOI: 10.1002/biot.202300390] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 02/21/2024]
Abstract
Organ-on-a-chip technology has shown great potential in disease modeling and drug evaluation. However, traditional organ-on-a-chip devices are mostly pump-dependent with low throughput, which makes it difficult to leverage their advantages. In this study, we have developed a generic, pump-free organ-on-a-chip platform consisting of a 32-unit chip and an adjustable rocker, facilitating high-throughput dynamic cell culture with straightforward operation. By utilizing the rocker to induce periodic fluid forces, we can achieve fluidic conditions similar to those obtained with traditional pump-based systems. Through constructing a gut-on-a-chip model, we observed remarkable enhancements in the expression of barrier-associated proteins and the spatial distribution of differentiated intestinal cells compared to static culture. Furthermore, RNA sequencing analysis unveiled enriched pathways associated with cell proliferation, lipid transport, and drug metabolism, indicating the ability of the platform to mimic critical physiological processes. Additionally, we tested seven drugs that represent a range of high, medium, and low in vivo permeability using this model and found a strong correlation between their Papp values and human Fa, demonstrating the capability of this model for drug absorption evaluation. Our findings highlight the potential of this pump-free organ-on-a-chip platform as a valuable tool for advancing drug development and enabling personalized medicine.
Collapse
Affiliation(s)
- Yaqiong Guo
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yingying Xie
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China
| |
Collapse
|
28
|
Young AT, Deal H, Rusch G, Pozdin VA, Brown AC, Daniele M. Simple design for membrane-free microphysiological systems to model the blood-tissue barriers. ORGANS-ON-A-CHIP 2023; 5:100032. [PMID: 39949484 PMCID: PMC11823427 DOI: 10.1016/j.ooc.2023.100032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/16/2025]
Abstract
Microphysiological systems (MPS) incorporate physiologically relevant microanatomy, mechanics, and cells to mimic tissue function. Reproducible and standardized in vitro models of tissue barriers, such as the blood-tissue interface (BTI), are critical for next-generation MPS applications in research and industry. Many models of the BTI are limited by the need for semipermeable membranes, use of homogenous cell populations, or 2D culture. These factors limit the relevant endothelial-epithelial contact and 3D transport, which would best mimic the BTI. Current models are also difficult to assemble, requiring precise alignment and layering of components. The work reported herein details the engineering of a BTI-on-a-chip (BTI Chip) that addresses current disadvantages by demonstrating a single layer, membrane-free design. Laminar flow profiles, photocurable hydrogel scaffolds, and human cell lines were used to construct a BTI Chip that juxtaposes an endothelium in direct contact with a 3D engineered tissue. A biomaterial composite, gelatin methacryloyl and 8-arm polyethylene glycol thiol, was used for in situ fabrication of a tissue structure within a Y-shaped microfluidic device. To produce the BTI, a laminar flow profile was achieved by flowing a photocurable precursor solution alongside phosphate buffered saline. Immediately after stopping flow, the scaffold underwent polymerization through a rapid exposure to UV light (<300 mJ/cm2). After scaffold formation, blood vessel endothelial cells were introduced and allowed to adhere directly to the 3D tissue scaffold, without barriers or phase guides. Fabrication of the BTI Chip was demonstrated in both an epithelial tissue model and blood-brain barrier (BBB) model. In the epithelial model, scaffolds were seeded with human dermal fibroblasts. For the BBB models, scaffolds were seeded with the immortalized glial cell line, SVGP12. The BTI Chip microanatomy was analyzed post facto by immunohistochemistry, showing the uniform production of a patent endothelium juxtaposed with a 3D engineered tissue. Fluorescent tracer molecules were used to characterize the permeability of the BTI Chip. The BTI Chips were challenged with an efflux pump inhibitor, cyclosporine A, to assess physiological function and endothelial cell activation. Operation of physiologically relevant BTI Chips and a novel means for high-throughput MPS generation was demonstrated, enabling future development for drug candidate screening and fundamental biological investigations.
Collapse
Affiliation(s)
- Ashlyn T. Young
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695, USA
| | - Halston Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695, USA
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC, 27606, USA
| | - Gabrielle Rusch
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695, USA
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC, 27606, USA
| | - Vladimir A. Pozdin
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL, USA
- Department of Mechanical & Materials Engineering, Florida International University, Miami, FL, USA
| | - Ashley C. Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695, USA
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC, 27606, USA
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695, USA
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC, 27606, USA
- Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh NC, 27695, USA
| |
Collapse
|
29
|
Young AT, Deal H, Rusch G, Pozdin VA, Brown AC, Daniele M. Simple Design for Membrane-Free Microphysiological Systems to Model the Blood-Tissue Barriers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563328. [PMID: 37961220 PMCID: PMC10634696 DOI: 10.1101/2023.10.20.563328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Microphysiological systems (MPS) incorporate physiologically relevant microanatomy, mechanics, and cells to mimic tissue function. Reproducible and standardized in vitro models of tissue barriers, such as the blood-tissue interface (BTI), are critical for next-generation MPS applications in research and industry. Many models of the BTI are limited by the need for semipermeable membranes, use of homogenous cell populations, or 2D culture. These factors limit the relevant endothelial-epithelial contact and 3D transport, which would best mimic the BTI. Current models are also difficult to assemble, requiring precise alignment and layering of components. The work reported herein details the engineering of a BTI-on-a-chip (BTI Chip) that addresses current disadvantages by demonstrating a single layer, membrane-free design. Laminar flow profiles, photocurable hydrogel scaffolds, and human cell lines were used to construct a BTI Chip that juxtaposes an endothelium in direct contact with a 3D engineered tissue. A biomaterial composite, gelatin methacryloyl and 8-arm polyethylene glycol thiol, was used for in situ fabrication of a tissue structure within a Y-shaped microfluidic device. To produce the BTI, a laminar flow profile was achieved by flowing a photocurable precursor solution alongside phosphate buffered saline. Immediately after stopping flow, the scaffold underwent polymerization through a rapid exposure to UV light (<300 mJ·cm-2). After scaffold formation, blood vessel endothelial cells were introduced and allowed to adhere directly to the 3D tissue scaffold, without barriers or phase guides. Fabrication of the BTI Chip was demonstrated in both an epithelial tissue model and blood-brain barrier (BBB) model. In the epithelial model, scaffolds were seeded with human dermal fibroblasts. For the BBB models, scaffolds were seeded with the immortalized glial cell line, SVGP12. The BTI Chip microanatomy was analyzed post facto by immunohistochemistry, showing the uniform production of a patent endothelium juxtaposed with a 3D engineered tissue. Fluorescent tracer molecules were used to characterize the permeability of the BTI Chip. The BTI Chips were challenged with an efflux pump inhibitor, cyclosporine A, to assess physiological function and endothelial cell activation. Operation of physiologically relevant BTI Chips and a novel means for high-throughput MPS generation was demonstrated, enabling future development for drug candidate screening and fundamental biological investigations.
Collapse
Affiliation(s)
- Ashlyn T. Young
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695 (USA)
| | - Halston Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695 (USA)
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Gabrielle Rusch
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695 (USA)
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Vladimir A. Pozdin
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL (USA)
- Department of Mechanical & Materials Engineering, Florida International University, Miami, FL (USA)
| | - Ashley C. Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695 (USA)
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh NC, 27695 (USA)
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
- Department of Mechanical & Materials Engineering, Florida International University, Miami, FL (USA)
| |
Collapse
|
30
|
Chen Y, Pan R, Mei L, Tian P, Wang L, Zhao J, Chen W, Wang G. Colon-Targeted Delivery of Indole Acetic Acid Helps Regulate Gut Motility by Activating the AHR Signaling Pathway. Nutrients 2023; 15:4282. [PMID: 37836566 PMCID: PMC10574622 DOI: 10.3390/nu15194282] [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: 09/15/2023] [Revised: 10/01/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Intestinal peristalsis is vital for gastrointestinal physiology and host homeostasis and is frequently dysregulated in intestinal disorders. Gut microbiota can regulate gut motility, especially through the tryptophan metabolism pathway. However, the role of indoles as microbial tryptophan metabolites in colonic function requires further exploration. Here, we show that the delivery of indole acetic acid (IAA) targeting the colon can improve gut motility by activating the aryl hydrocarbon receptor (AHR). To achieve colon-targeted delivery, Eudragit S-100 (ES) and chitosan (CS) were used as drug carriers. After optimisation, IAA-loaded ES-coated CS nanoparticles exhibited an encapsulation efficiency of 83% and a drug-loading capacity of 16%. These nanoparticles exhibited pH-dependent characteristics and remained stable in acidic conditions and the upper intestine. In simulated intestinal fluid (pH 7.4) and colonic lumen, considerable amounts of IAA were released after approximately 4 h. Compared with free IAA, the nanoparticles exerted enhanced therapeutic effects on gut movement disorders induced by loperamide. The efficacy of IAA treatment was attributable to the activation of the AHR signalling pathway and increased levels of AHR agonists. Furthermore, the oral administration of IAA-loaded nanoparticles promoted serotonin secretion and maintained the intestinal barrier function. The experimental outcomes demonstrate the efficiency of the proposed colon-specific delivery system and highlight the role of IAA, produced by gut microbiota metabolism, in regulating gut peristalsis through AHR activation.
Collapse
Affiliation(s)
- Ying Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ruili Pan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Liya Mei
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Linlin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.C.); (R.P.); (L.M.); (P.T.); (L.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| |
Collapse
|
31
|
Pedroza Matute S, Iyavoo S. Exploring the gut microbiota: lifestyle choices, disease associations, and personal genomics. Front Nutr 2023; 10:1225120. [PMID: 37867494 PMCID: PMC10585655 DOI: 10.3389/fnut.2023.1225120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
The gut microbiota is a rich and dynamic ecosystem that actively interacts with the human body, playing a significant role in the state of health and disease of the host. Diet, exercise, mental health, and other factors have exhibited the ability to influence the gut bacterial composition, leading to changes that can prevent and improve, or favor and worsen, both intestinal and extra-intestinal conditions. Altered gut microbial states, or 'dysbiosis', associated with conditions and diseases are often characterized by shifts in bacterial abundance and diversity, including an impaired Firmicutes to Bacteroidetes ratio. By understanding the effect of lifestyle on the gut microbiota, personalized advice can be generated to suit each individual profile and foster the adoption of lifestyle changes that can both prevent and ameliorate dysbiosis. The delivery of effective and reliable advice, however, depends not only on the available research and current understanding of the topic, but also on the methods used to assess individuals and to discover the associations, which can introduce bias at multiple stages. The aim of this review is to summarize how human gut microbial variability is defined and what lifestyle choices and diseases have shown association with gut bacterial composition. Furthermore, popular methods to investigate the human gut microbiota are outlined, with a focus on the possible bias caused by the lack of use of standardized methods. Finally, an overview of the current state of personalized advice based on gut microbiota testing is presented, underlining its power and limitations.
Collapse
Affiliation(s)
| | - Sasitaran Iyavoo
- Nkaarco Diagnostics Limited, Norwich, United Kingdom
- School of Chemistry, College of Health and Science, University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
32
|
Aizpurua O, Blijleven K, Trivedi U, Gilbert MTP, Alberdi A. Unravelling animal-microbiota evolution on a chip. Trends Microbiol 2023; 31:995-1002. [PMID: 37217368 DOI: 10.1016/j.tim.2023.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Whether and how microorganisms have shaped the evolution of their animal hosts is a major question in biology. Although many animal evolutionary processes appear to correlate with changes in their associated microbial communities, the mechanistic processes leading to these patterns and their causal relationships are still far from being resolved. Gut-on-a-chip models provide an innovative approach that expands beyond the potential of conventional microbiome profiling to study how different animals sense and react to microbes by comparing responses of animal intestinal tissue models to different microbial stimuli. This complementary knowledge can contribute to our understanding of how host genetic features facilitate or prevent different microbiomes from being assembled, and in doing so elucidate the role of host-microbiota interactions in animal evolution.
Collapse
Affiliation(s)
- Ostaizka Aizpurua
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Kees Blijleven
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Urvish Trivedi
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - M Thomas P Gilbert
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark; University Museum, NTNU, Trondheim, Norway
| | - Antton Alberdi
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Nelson MT, Coia HG, Holt C, Greenwood ES, Narayanan L, Robinson PJ, Merrill EA, Litteral V, Goodson MS, Saldanha RJ, Grogg MW, Mauzy CA. Evaluation of Human Performance Aiding Live Synthetically Engineered Bacteria in a Gut-on-a-Chip. ACS Biomater Sci Eng 2023; 9:5136-5150. [PMID: 36198112 DOI: 10.1021/acsbiomaterials.2c00774] [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] [Indexed: 11/30/2022]
Abstract
Synbiotics are a new class of live therapeutics employing engineered genetic circuits. The rapid adoption of genetic editing tools has catalyzed the expansion of possible synbiotics, exceeding traditional testing paradigms in terms of both throughput and model complexity. Herein, we present a simplistic gut-chip model using common Caco2 and HT-29 cell lines to establish a dynamic human screening platform for a cortisol sensing tryptamine producing synbiotic for cognitive performance sustainment. The synbiotic, SYN, was engineered from the common probiotic E. coli Nissle 1917 strain. It had the ability to sense cortisol at physiological concentrations, resulting in the activation of a genetic circuit that produces tryptophan decarboxylase and converts bioavailable tryptophan to tryptamine. SYN was successfully cultivated within the gut-chip showing log-phase growth comparable to the wild-type strain. Tryptophan metabolism occurred quickly in the gut compartment when exposed to 5 μM cortisol, resulting in the complete conversion of bioavailable tryptophan into tryptamine. The flux of tryptophan and tryptamine from the gut to the vascular compartment of the chip was delayed by 12 h, as indicated by the detectable tryptamine in the vascular compartment. The gut-chip provided a stable environment to characterize the sensitivity of the cortisol sensor and dynamic range by altering cortisol and tryptophan dosimetry. Collectively, the human gut-chip provided human relevant apparent permeability to assess tryptophan and tryptamine metabolism, production, and transport, enabled host analyses of cellular viability and pro-inflammatory cytokine secretion, and succeeded in providing an efficacy test of a novel synbiotic. Organ-on-a-chip technology holds promise in aiding traditional therapeutic pipelines to more rapidly down select high potential compounds that reduce the failure rate and accelerate the opportunity for clinical intervention.
Collapse
Affiliation(s)
- M Tyler Nelson
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| | - Heidi G Coia
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
- National Research Council, The National Academies of Sciences, Engineering, and Medicine, 500 Fifth Street N.W., Washington, D.C. 20001, United States
| | - Corey Holt
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| | - Eric S Greenwood
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
- Oak Ridge Institute for Science and Education, 1299 Bethel Valley Road, Oak Ridge, Tennessee 37830, United States
| | - Latha Narayanan
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
- The Henry M. Jackson Foundation, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Peter J Robinson
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
- The Henry M. Jackson Foundation, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Elaine A Merrill
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| | - Vaughn Litteral
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
- UES Inc., 4401 Dayton-Xenia Road, Dayton, Ohio 45432, United States
| | - Michael S Goodson
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| | - Roland J Saldanha
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| | - Matthew W Grogg
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| | - Camilla A Mauzy
- United States Air Force Research Laboratory, 711th Human Performance Wing, 2510 N 5th Street, Bldg. 840, Wright-Patterson AFB, Ohio 45433, United States
| |
Collapse
|
34
|
Pasquereau-Kotula E, Nigro G, Dingli F, Loew D, Poullet P, Xu Y, Kopetz S, Davis J, Peduto L, Robbe-Masselot C, Sansonetti P, Trieu-Cuot P, Dramsi S. Global proteomic identifies multiple cancer-related signaling pathways altered by a gut pathobiont associated with colorectal cancer. Sci Rep 2023; 13:14960. [PMID: 37696912 PMCID: PMC10495336 DOI: 10.1038/s41598-023-41951-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
In this work, we investigated the oncogenic role of Streptococcus gallolyticus subsp. gallolyticus (SGG), a gut bacterium associated with colorectal cancer (CRC). We showed that SGG UCN34 accelerates colon tumor development in a chemically induced CRC murine model. Full proteome and phosphoproteome analysis of murine colons chronically colonized by SGG UCN34 revealed that 164 proteins and 725 phosphorylation sites were differentially regulated. Ingenuity Pathway Analysis (IPA) indicates a pro-tumoral shift specifically induced by SGG UCN34, as ~ 90% of proteins and phosphoproteins identified were associated with digestive cancer. Comprehensive analysis of the altered phosphoproteins using ROMA software revealed up-regulation of several cancer hallmark pathways such as MAPK, mTOR and integrin/ILK/actin, affecting epithelial and stromal colonic cells. Importantly, an independent analysis of protein arrays of human colon tumors colonized with SGG showed up-regulation of PI3K/Akt/mTOR and MAPK pathways, providing clinical relevance to our findings. To test SGG's capacity to induce pre-cancerous transformation of the murine colonic epithelium, we grew ex vivo organoids which revealed unusual structures with compact morphology. Taken together, our results demonstrate the oncogenic role of SGG UCN34 in a murine model of CRC associated with activation of multiple cancer-related signaling pathways.
Collapse
Affiliation(s)
- Ewa Pasquereau-Kotula
- Biology of Gram-Positive Pathogens Unit, Institut Pasteur, Université Paris Cité, CNRS UMR6047, 75015, Paris, France.
| | - Giulia Nigro
- Stroma, Inflammation and Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, 75015, Paris, France
- Microenvironment and Immunity Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, 75015, Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, CurieCoreTech Spectrométrie de Masse Protéomique, 75005, Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, CurieCoreTech Spectrométrie de Masse Protéomique, 75005, Paris, France
| | - Patrick Poullet
- Institut Curie, Bioinformatics Core Facility (CUBIC), INSERM U900, PSL Research University, Mines Paris Tech, 75005, Paris, France
| | - Yi Xu
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Bryan, TX, USA
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Davis
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- University of Kansas, Kansas City, KS, USA
| | - Lucie Peduto
- Stroma, Inflammation and Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, 75015, Paris, France
| | - Catherine Robbe-Masselot
- Université de Lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Philippe Sansonetti
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, and College de France, 75005, Paris, France
| | - Patrick Trieu-Cuot
- Biology of Gram-Positive Pathogens Unit, Institut Pasteur, Université Paris Cité, CNRS UMR6047, 75015, Paris, France
| | - Shaynoor Dramsi
- Biology of Gram-Positive Pathogens Unit, Institut Pasteur, Université Paris Cité, CNRS UMR6047, 75015, Paris, France.
| |
Collapse
|
35
|
Reddy N, Lynch B, Gujral J, Karnik K. Alternatives to animal testing in toxicity testing: Current status and future perspectives in food safety assessments. Food Chem Toxicol 2023; 179:113944. [PMID: 37453475 DOI: 10.1016/j.fct.2023.113944] [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: 04/20/2023] [Revised: 06/29/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
The development of alternative methods to animal testing has gained great momentum since Russel and Burch introduced the "3Rs" concept of Reduction, Refinement, and Replacement of animals in safety testing in 1959. Several alternatives to animal testing have since been introduced, including but not limited to in vitro and in chemico test systems, in silico models, and computational models (e.g., [quantitative] structural activity relationship models, high-throughput screens, organ-on-chip models, and genomics or bioinformatics) to predict chemical toxicity. Furthermore, several agencies have developed robust integrated testing strategies to determine chemical toxicity. The cosmetics sector is pioneering the adoption of alternative methodologies for safety evaluations, and other sectors are aiming to completely abandon animal testing by 2035. However, beyond the use of in vitro genetic testing, agencies regulating the food industry have been slow to implement alternative methodologies into safety evaluations compared with other sectors; setting health-based guidance values for food ingredients requires data from systemic toxicity, and to date, no standalone validated alternative models to assess systemic toxicity exist. The abovementioned models show promise for assessing systemic toxicity with further research. In this paper, we review the current alternatives and their applicability and limitations in food safety evaluations.
Collapse
Affiliation(s)
- Navya Reddy
- Intertek Health Sciences Inc., 2233 Argentia Rd, Suite 201, Mississauga, ON, L5N 2X7, Canada
| | - Barry Lynch
- Intertek Health Sciences Inc., 2233 Argentia Rd, Suite 201, Mississauga, ON, L5N 2X7, Canada.
| | - Jaspreet Gujral
- Tate & Lyle, 5450 Prairie Stone Pkwy, Hoffman Estates, IL, 60192, USA
| | - Kavita Karnik
- Tate & Lyle PLC, 5 Marble Arch, London, W1H 7EJ, United Kingdom
| |
Collapse
|
36
|
Donnaloja F, Izzo L, Campanile M, Perottoni S, Boeri L, Fanizza F, Sardelli L, Jacchetti E, Raimondi MT, Rito LD, Craparotta I, Bolis M, Giordano C, Albani D. Human gut epithelium features recapitulated in MINERVA 2.0 millifluidic organ-on-a-chip device. APL Bioeng 2023; 7:036117. [PMID: 37736017 PMCID: PMC10511260 DOI: 10.1063/5.0144862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023] Open
Abstract
We developed an innovative millifluidic organ-on-a-chip device, named MINERVA 2.0, that is optically accessible and suitable to serial connection. In the present work, we evaluated MINERVA 2.0 as millifluidic gut epithelium-on-a-chip by using computational modeling and biological assessment. We also tested MINERVA 2.0 in a serially connected configuration prodromal to address the complexity of multiorgan interaction. Once cultured under perfusion in our device, human gut immortalized Caco-2 epithelial cells were able to survive at least up to 7 days and form a three-dimensional layer with detectable tight junctions (occludin and zonulin-1 positive). Functional layer development was supported by measurable trans-epithelial resistance and FITC-dextran permeability regulation, together with mucin-2 expression. The dynamic culturing led to a specific transcriptomic profile, assessed by RNASeq, with a total of 524 dysregulated transcripts (191 upregulated and 333 downregulated) between static and dynamic condition. Overall, the collected results suggest that our gut-on-a-chip millifluidic model displays key gut epithelium features and, thanks to its modular design, may be the basis to build a customizable multiorgan-on-a-chip platform.
Collapse
Affiliation(s)
- Francesca Donnaloja
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Marzia Campanile
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Francesca Fanizza
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Lorenzo Sardelli
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Manuela T. Raimondi
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Laura Di Rito
- Department of Oncology, Computational Oncology Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Craparotta
- Department of Oncology, Computational Oncology Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Bolis
- Department of Oncology, Computational Oncology Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
37
|
Yang W, Lipert M, Nofsinger R. Current screening, design, and delivery approaches to address low permeability of chemically synthesized modalities in drug discovery and early clinical development. Drug Discov Today 2023; 28:103685. [PMID: 37356613 DOI: 10.1016/j.drudis.2023.103685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/09/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
A drug's permeability across biological membranes is a key property associated with the successful development of an orally absorbed drug candidate. Although a variety of methods are available for predicting and assessing permeability, some are more preferred than others at specific stages of drug discovery and development across the pharmaceutical industry. Permeability measurements may be interpreted differently depending on the chosen method. Herein, we present a refreshed perspective on the screening approaches and philosophy in permeability evaluation, from early drug discovery to early clinical development. Additionally, we review and discuss chemical design and drug delivery technologies that can be leveraged to overcome permeability challenges, which are increasingly being used with emerging modalities.
Collapse
Affiliation(s)
- Wenzhan Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA.
| | - Maya Lipert
- Molecular Profiling and Drug Delivery, Small Molecule CMC Development, AbbVie, Inc., North Chicago, IL, USA
| | | |
Collapse
|
38
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
39
|
Corral-Nájera K, Chauhan G, Serna-Saldívar SO, Martínez-Chapa SO, Aeinehvand MM. Polymeric and biological membranes for organ-on-a-chip devices. MICROSYSTEMS & NANOENGINEERING 2023; 9:107. [PMID: 37649779 PMCID: PMC10462672 DOI: 10.1038/s41378-023-00579-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 05/18/2023] [Accepted: 06/20/2023] [Indexed: 09/01/2023]
Abstract
Membranes are fundamental elements within organ-on-a-chip (OOC) platforms, as they provide adherent cells with support, allow nutrients (and other relevant molecules) to permeate/exchange through membrane pores, and enable the delivery of mechanical or chemical stimuli. Through OOC platforms, physiological processes can be studied in vitro, whereas OOC membranes broaden knowledge of how mechanical and chemical cues affect cells and organs. OOCs with membranes are in vitro microfluidic models that are used to replace animal testing for various applications, such as drug discovery and disease modeling. In this review, the relevance of OOCs with membranes is discussed as well as their scaffold and actuation roles, properties (physical and material), and fabrication methods in different organ models. The purpose was to aid readers with membrane selection for the development of OOCs with specific applications in the fields of mechanistic, pathological, and drug testing studies. Mechanical stimulation from liquid flow and cyclic strain, as well as their effects on the cell's increased physiological relevance (IPR), are described in the first section. The review also contains methods to fabricate synthetic and ECM (extracellular matrix) protein membranes, their characteristics (e.g., thickness and porosity, which can be adjusted depending on the application, as shown in the graphical abstract), and the biological materials used for their coatings. The discussion section joins and describes the roles of membranes for different research purposes and their advantages and challenges.
Collapse
Affiliation(s)
- Kendra Corral-Nájera
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Gaurav Chauhan
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Sergio O. Serna-Saldívar
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Sergio O. Martínez-Chapa
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| | - Mohammad Mahdi Aeinehvand
- School of Engineering and Science, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, 64849 Mexico
| |
Collapse
|
40
|
Kim J, Kim J, Jin Y, Cho SW. In situbiosensing technologies for an organ-on-a-chip. Biofabrication 2023; 15:042002. [PMID: 37587753 DOI: 10.1088/1758-5090/aceaae] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Thein vitrosimulation of organs resolves the accuracy, ethical, and cost challenges accompanyingin vivoexperiments. Organoids and organs-on-chips have been developed to model thein vitro, real-time biological and physiological features of organs. Numerous studies have deployed these systems to assess thein vitro, real-time responses of an organ to external stimuli. Particularly, organs-on-chips can be most efficiently employed in pharmaceutical drug development to predict the responses of organs before approving such drugs. Furthermore, multi-organ-on-a-chip systems facilitate the close representations of thein vivoenvironment. In this review, we discuss the biosensing technology that facilitates thein situ, real-time measurements of organ responses as readouts on organ-on-a-chip systems, including multi-organ models. Notably, a human-on-a-chip system integrated with automated multi-sensing will be established by further advancing the development of chips, as well as their assessment techniques.
Collapse
Affiliation(s)
- Jinyoung Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Junghoon Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Institute for Basic Science (IBS), Center for Nanomedicine, Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
41
|
Masloh S, Culot M, Gosselet F, Chevrel A, Scapozza L, Zeisser Labouebe M. Challenges and Opportunities in the Oral Delivery of Recombinant Biologics. Pharmaceutics 2023; 15:pharmaceutics15051415. [PMID: 37242657 DOI: 10.3390/pharmaceutics15051415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Recombinant biological molecules are at the cutting-edge of biomedical research thanks to the significant progress made in biotechnology and a better understanding of subcellular processes implicated in several diseases. Given their ability to induce a potent response, these molecules are becoming the drugs of choice for multiple pathologies. However, unlike conventional drugs which are mostly ingested, the majority of biologics are currently administered parenterally. Therefore, to improve their limited bioavailability when delivered orally, the scientific community has devoted tremendous efforts to develop accurate cell- and tissue-based models that allow for the determination of their capacity to cross the intestinal mucosa. Furthermore, several promising approaches have been imagined to enhance the intestinal permeability and stability of recombinant biological molecules. This review summarizes the main physiological barriers to the oral delivery of biologics. Several preclinical in vitro and ex vivo models currently used to assess permeability are also presented. Finally, the multiple strategies explored to address the challenges of administering biotherapeutics orally are described.
Collapse
Affiliation(s)
- Solene Masloh
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Maxime Culot
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Anne Chevrel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Magali Zeisser Labouebe
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| |
Collapse
|
42
|
Qi P, Lv J, Yan X, Bai L, Zhang L. Microfluidics: Insights into Intestinal Microorganisms. Microorganisms 2023; 11:1134. [PMID: 37317109 DOI: 10.3390/microorganisms11051134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 06/16/2023] Open
Abstract
Microfluidics is a system involving the treatment or manipulation of microscale (10-9 to 10-18 L) fluids using microchannels (10 to 100 μm) contained on a microfluidic chip. Among the different methodologies used to study intestinal microorganisms, new methods based on microfluidic technology have been receiving increasing attention in recent years. The intestinal tracts of animals are populated by a vast array of microorganisms that have been established to play diverse functional roles beneficial to host physiology. This review is the first comprehensive coverage of the application of microfluidics technology in intestinal microbial research. In this review, we present a brief history of microfluidics technology and describe its applications in gut microbiome research, with a specific emphasis on the microfluidic technology-based intestine-on-a-chip, and also discuss the advantages and application prospects of microfluidic drug delivery systems in intestinal microbial research.
Collapse
Affiliation(s)
- Ping Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jin Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiangdong Yan
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Liuhui Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
43
|
Chen H, Luo Z, Lin X, Zhu Y, Zhao Y. Sensors-integrated organ-on-a-chip for biomedical applications. NANO RESEARCH 2023; 16:1-28. [PMID: 37359077 PMCID: PMC10130312 DOI: 10.1007/s12274-023-5651-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 06/28/2023]
Abstract
As a promising new micro-physiological system, organ-on-a-chip has been widely utilized for in vitro pharmaceutical study and tissues engineering based on the three-dimensional constructions of tissues/organs and delicate replication of in vivo-like microenvironment. To better observe the biological processes, a variety of sensors have been integrated to realize in-situ, real-time, and sensitive monitoring of critical signals for organs development and disease modeling. Herein, we discuss the recent research advances made with respect to sensors-integrated organ-on-a-chip in this overall review. Firstly, we briefly explore the underlying fabrication procedures of sensors within microfluidic platforms and several classifications of sensory principles. Then, emphasis is put on the highlighted applications of different types of organ-on-a-chip incorporated with various sensors. Last but not least, perspective on the remaining challenges and future development of sensors-integrated organ-on-a-chip are presented.
Collapse
Affiliation(s)
- Hanxu Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Zhiqiang Luo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Yujuan Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001 China
| |
Collapse
|
44
|
Liu S, Kumari S, He H, Mishra P, Singh BN, Singh D, Liu S, Srivastava P, Li C. Biosensors integrated 3D organoid/organ-on-a-chip system: A real-time biomechanical, biophysical, and biochemical monitoring and characterization. Biosens Bioelectron 2023; 231:115285. [PMID: 37058958 DOI: 10.1016/j.bios.2023.115285] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
As a full-fidelity simulation of human cells, tissues, organs, and even systems at the microscopic scale, Organ-on-a-Chip (OOC) has significant ethical advantages and development potential compared to animal experiments. The need for the design of new drug high-throughput screening platforms and the mechanistic study of human tissues/organs under pathological conditions, the evolving advances in 3D cell biology and engineering, etc., have promoted the updating of technologies in this field, such as the iteration of chip materials and 3D printing, which in turn facilitate the connection of complex multi-organs-on-chips for simulation and the further development of technology-composite new drug high-throughput screening platforms. As the most critical part of organ-on-a-chip design and practical application, verifying the success of organ model modeling, i.e., evaluating various biochemical and physical parameters in OOC devices, is crucial. Therefore, this paper provides a logical and comprehensive review and discussion of the advances in organ-on-a-chip detection and evaluation technologies from a broad perspective, covering the directions of tissue engineering scaffolds, microenvironment, single/multi-organ function, and stimulus-based evaluation, and provides a more comprehensive review of the progress in the significant organ-on-a-chip research areas in the physiological state.
Collapse
Affiliation(s)
- Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Shikha Kumari
- School of Biochemical Engineering, IIT BHU, Varanasi, Uttar Pradesh, India
| | - Hongyi He
- West China School of Medicine & West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Parichita Mishra
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Bhisham Narayan Singh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Divakar Singh
- School of Biochemical Engineering, IIT BHU, Varanasi, Uttar Pradesh, India
| | - Sutong Liu
- Juxing College of Digital Economics, Haikou University of Economics, Haikou, 570100, China
| | - Pradeep Srivastava
- School of Biochemical Engineering, IIT BHU, Varanasi, Uttar Pradesh, India.
| | - Chenzhong Li
- Biomedical Engineering, School of Medicine, The Chinese University of Hong Kong(Shenzhen), Shenzhen, 518172, China.
| |
Collapse
|
45
|
Li ZA, Sant S, Cho SK, Goodman SB, Bunnell BA, Tuan RS, Gold MS, Lin H. Synovial joint-on-a-chip for modeling arthritis: progress, pitfalls, and potential. Trends Biotechnol 2023; 41:511-527. [PMID: 35995600 PMCID: PMC9938846 DOI: 10.1016/j.tibtech.2022.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 12/30/2022]
Abstract
Disorders of the synovial joint, such as osteoarthritis (OA) and rheumatoid arthritis (RA), afflict a substantial proportion of the global population. However, current clinical management has not been focused on fully restoring the native function of joints. Organ-on-chip (OoC), also called a microphysiological system, which typically accommodates multiple human cell-derived tissues/organs under physiological culture conditions, is an emerging platform that potentially overcomes the limitations of current models in developing therapeutics. Herein, we review major steps in the generation of OoCs for studying arthritis, discuss the challenges faced when these novel platforms enter the next phase of development and application, and present the potential for OoC technology to investigate the pathogenesis of joint diseases and the development of efficacious therapies.
Collapse
Affiliation(s)
- Zhong Alan Li
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Sung Kwon Cho
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Stuart B Goodman
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rocky S Tuan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, China
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
46
|
Biosensor integrated brain-on-a-chip platforms: Progress and prospects in clinical translation. Biosens Bioelectron 2023; 225:115100. [PMID: 36709589 DOI: 10.1016/j.bios.2023.115100] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/07/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
Because of the brain's complexity, developing effective treatments for neurological disorders is a formidable challenge. Research efforts to this end are advancing as in vitro systems have reached the point that they can imitate critical components of the brain's structure and function. Brain-on-a-chip (BoC) was first used for microfluidics-based systems with small synthetic tissues but has expanded recently to include in vitro simulation of the central nervous system (CNS). Defining the system's qualifying parameters may improve the BoC for the next generation of in vitro platforms. These parameters show how well a given platform solves the problems unique to in vitro CNS modeling (like recreating the brain's microenvironment and including essential parts like the blood-brain barrier (BBB)) and how much more value it offers than traditional cell culture systems. This review provides an overview of the practical concerns of creating and deploying BoC systems and elaborates on how these technologies might be used. Not only how advanced biosensing technologies could be integrated with BoC system but also how novel approaches will automate assays and improve point-of-care (PoC) diagnostics and accurate quantitative analyses are discussed. Key challenges providing opportunities for clinical translation of BoC in neurodegenerative disorders are also addressed.
Collapse
|
47
|
Marrero D, Guimera A, Maes L, Villa R, Alvarez M, Illa X. Organ-on-a-chip with integrated semitransparent organic electrodes for barrier function monitoring. LAB ON A CHIP 2023; 23:1825-1834. [PMID: 36810654 DOI: 10.1039/d2lc01097f] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Organs-on-a-chip (OoC) are cell culture platforms that replicate key functional units of tissues in vitro. Barrier integrity and permeability evaluation are of utmost importance when studying barrier-forming tissues. Impedance spectroscopy is a powerful tool and is widely used to monitor barrier permeability and integrity in real-time. However, data comparison across devices is misleading due to the generation of a non-homogenous field across the tissue barrier, making impedance data normalization very challenging. In this work, we address this issue by integrating PEDOT:PSS electrodes for barrier function monitoring with impedance spectroscopy. The semitransparent PEDOT:PSS electrodes cover the entire cell culture membrane providing a homogenous electric field across the entire membrane making the cell culture area equally accountable to the measured impedance. To the best of our knowledge, PEDOT:PSS has never been used solely to monitor the impedance of cellular barriers while enabling optical inspection in the OoC. The performance of the device is demonstrated by lining the device with intestinal cells where we monitored barrier formation under flow conditions, as well as barrier disruption and recovery under exposure to a permeability enhancer. The barrier tightness and integrity, and the intercellular cleft have been evaluated by analyzing the full impedance spectrum. Furthermore, the device is autoclavable paving the way toward more sustainable OoC options.
Collapse
Affiliation(s)
- Denise Marrero
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Campus UAB, Bellaterra, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y Nanomedicina, Madrid, 50018, Spain
| | - Anton Guimera
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Campus UAB, Bellaterra, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y Nanomedicina, Madrid, 50018, Spain
| | - Laure Maes
- Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Ghent Gut Inflammation Group, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Gent, Belgium
| | - Rosa Villa
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Campus UAB, Bellaterra, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y Nanomedicina, Madrid, 50018, Spain
| | - Mar Alvarez
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Campus UAB, Bellaterra, 08193, Barcelona, Spain.
| | - Xavi Illa
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Campus UAB, Bellaterra, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y Nanomedicina, Madrid, 50018, Spain
| |
Collapse
|
48
|
Dai M, Xiao G, Shao M, Zhang YS. The Synergy between Deep Learning and Organs-on-Chips for High-Throughput Drug Screening: A Review. BIOSENSORS 2023; 13:389. [PMID: 36979601 PMCID: PMC10046732 DOI: 10.3390/bios13030389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Organs-on-chips (OoCs) are miniature microfluidic systems that have arguably become a class of advanced in vitro models. Deep learning, as an emerging topic in machine learning, has the ability to extract a hidden statistical relationship from the input data. Recently, these two areas have become integrated to achieve synergy for accelerating drug screening. This review provides a brief description of the basic concepts of deep learning used in OoCs and exemplifies the successful use cases for different types of OoCs. These microfluidic chips are of potential to be assembled as highly potent human-on-chips with complex physiological or pathological functions. Finally, we discuss the future supply with perspectives and potential challenges in terms of combining OoCs and deep learning for image processing and automation designs.
Collapse
Affiliation(s)
- Manna Dai
- College of Physics and Information Engineering, Fuzhou University, Fuzhou 350108, China
- Computing and Intelligence Department, Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, #16-16 Connexis, Singapore 138632, Singapore
| | - Gao Xiao
- College of Environment and Safety Engineering, Fuzhou University, Fuzhou 350108, China
- Department of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Ming Shao
- Department of Computer and Information Science, College of Engineering, University of Massachusetts Dartmouth, North Dartmouth, MA 02747, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
49
|
Xiang X, Wang X, Shang Y, Ding Y. Microfluidic intestine-on-a-chip: Current progress and further perspectives of probiotic-foodborne pathogen interactions. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
50
|
Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G, Maruccio G. Organs-on-chips technologies – A guide from disease models to opportunities for drug development. Biosens Bioelectron 2023; 231:115271. [PMID: 37060819 DOI: 10.1016/j.bios.2023.115271] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 11/24/2022] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Current in-vitro 2D cultures and animal models present severe limitations in recapitulating human physiopathology with striking discrepancies in estimating drug efficacy and side effects when compared to human trials. For these reasons, microphysiological systems, organ-on-chip and multiorgans microdevices attracted considerable attention as novel tools for high-throughput and high-content research to achieve an improved understanding of diseases and to accelerate the drug development process towards more precise and eventually personalized standards. This review takes the form of a guide on this fast-growing field, providing useful introduction to major themes and indications for further readings. We start analyzing Organs-on-chips (OOC) technologies for testing the major drug administration routes: (1) oral/rectal route by intestine-on-a-chip, (2) inhalation by lung-on-a-chip, (3) transdermal by skin-on-a-chip and (4) intravenous through vascularization models, considering how drugs penetrate in the bloodstream and are conveyed to their targets. Then, we focus on OOC models for (other) specific organs and diseases: (1) neurodegenerative diseases with brain models and blood brain barriers, (2) tumor models including their vascularization, organoids/spheroids, engineering and screening of antitumor drugs, (3) liver/kidney on chips and multiorgan models for gastrointestinal diseases and metabolic assessment of drugs and (4) biomechanical systems recapitulating heart, muscles and bones structures and related diseases. Successively, we discuss technologies and materials for organ on chips, analyzing (1) microfluidic tools for organs-on-chips, (2) sensor integration for real-time monitoring, (3) materials and (4) cell lines for organs on chips. (Nano)delivery approaches for therapeutics and their on chip assessment are also described. Finally, we conclude with a critical discussion on current significance/relevance, trends, limitations, challenges and future prospects in terms of revolutionary impact on biomedical research, preclinical models and drug development.
Collapse
Affiliation(s)
- Anna Grazia Monteduro
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Silvia Rizzato
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Giusi Caragnano
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Giuseppe Maruccio
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy.
| |
Collapse
|