1
|
Sakolish C, Moyer HL, Tsai HHD, Ford LC, Dickey AN, Bajaj P, Villenave R, Hewitt P, Ferguson SS, Stanko J, Rusyn I. Comparative analysis of the physiological and transport functions of various sources of renal proximal tubule cells under static and fluidic conditions in PhysioMimix T12 platform. Drug Metab Dispos 2025; 53:100001. [PMID: 39884810 DOI: 10.1124/dmd.124.001488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/16/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024] Open
Abstract
In vitro models that can faithfully replicate critical aspects of kidney tubule function such as directional drug transport are in high demand in pharmacology and toxicology. Accordingly, development and validation of new models is underway. The objective of this study was to characterize physiologic and transport functions of various sources of human renal proximal tubule epithelial cells (RPTECs). We tested telomerase reverse transcriptase 1 (TERT1)-immortalized RPTECs, including organic anion transporter 1 (OAT1)-, organic cation transporter 2 (OCT2)-, or OAT3-overexpressing variants and primary RPTECs. Cells were cultured on transwell membranes in static (24-well transwells) and fluidic (transwells in PhysioMimix T12 organ-on-chip with 2 μL/s flow) conditions. Barrier formation, transport, and gene expression were evaluated. We show that 2 commercially available primary RPTECs were not suitable for studies of directional transport on transwells because they formed a substandard barrier even though they exhibited higher expression of transporters, especially under flow. TERT1-parent, -OAT1, and -OAT3 cells formed robust barriers but were unaffected by flow. TERT1-OAT1 cells exhibited inhibitable para-aminohippurate transport that was enhanced by flow. However, efficient tenofovir secretion and perfluorooctanoic acid reabsorption by TERT1-OAT1 cells were not modulated by flow. Gene expression showed that TERT1 and TERT1-OAT1 cells were more correlated with human kidney than other cell lines but that flow did not have noticeable effects. Overall, our data show that addition of flow to in vitro studies of the renal proximal tubule may afford benefits in some aspects of modeling kidney function but that careful consideration of the impact such adaptations would have on the cost and throughput of the experiments is needed. SIGNIFICANCE STATEMENT: The topic of reproducibility and robustness of complex microphysiological systems is looming large in the field of biomedical research; therefore, uptake of these new models by the end-users is slow. This study systematically compared various renal proximal tubule epithelial cell sources and experimental conditions, aiming to identify the level of model complexity needed for testing renal tubule transport. We demonstrate that although tissue chips may afford some benefits, their throughput and complexity need careful consideration in each context of use.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Haley L Moyer
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Han-Hsuan D Tsai
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Lucie C Ford
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Allison N Dickey
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, Massachusetts
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck KGaA, Darmstadt, Germany
| | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Jason Stanko
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| |
Collapse
|
2
|
Yan S, Lu Y, An C, Hu W, Chen Y, Li Z, Wei W, Chen Z, Zeng X, Xu W, Lv Z, Pan F, Gao W, Wu Y. Biomechanical research using advanced micro-nano devices: In-Vitro cell Characterization focus. J Adv Res 2024:S2090-1232(24)00602-7. [PMID: 39701378 DOI: 10.1016/j.jare.2024.12.024] [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: 08/18/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cells in the body reside in a dynamic microenvironment subjected to various physical stimuli, where mechanical stimulation plays a crucial role in regulating cellular physiological behaviors and functions. AIM OF REVIEW Investigating the mechanisms and interactions of mechanical transmission is essential for understanding the physiological and functional interplay between cells and physical stimuli. Therefore, establishing an in vitro biomechanical stimulation cell culture system holds significant importance for research related to cellular biomechanics. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we primarily focused on various biomechanically relevant cell culture systems and highlighted the advancements and prospects in their preparation processes. Firstly, we discussed the types and characteristics of biomechanics present in the microenvironment within the human body. Subsequently, we introduced the research progress, working principles, preparation processes, potential advantages, applications, and challenges of various biomechanically relevant in vitro cell culture systems. Additionally, we summarized and categorized currently commercialized biomechanically relevant cell culture systems, offering a comprehensive reference for researchers in related fields.
Collapse
Affiliation(s)
- Shiqiang Yan
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China; Center of Cancer Immunology, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yan Lu
- Department of Otolaryngology Head & Neck Surgery, The First Hospital, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Changming An
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wanglai Hu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Yaofeng Chen
- Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Ziwen Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Wenbo Wei
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| | - Zongzheng Chen
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| | - Xianhai Zeng
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
| | - Wei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, Shandong, China
| | - Zhenghua Lv
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, Shandong, China.
| | - Fan Pan
- Center of Cancer Immunology, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China.
| | - Wei Gao
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China.
| | - Yongyan Wu
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China; Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
3
|
Kim MH, Lee Y, Seo GM, Park S. Advancements in Kidney-on-Chip: Antibiotic-Induced Kidney Injury and Future Directions. BIOCHIP JOURNAL 2024; 18:535-545. [DOI: 10.1007/s13206-024-00160-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 01/06/2025]
|
4
|
Çam SB, Çiftci E, Gürbüz N, Altun B, Korkusuz P. Allogeneic bone marrow mesenchymal stem cell-derived exosomes alleviate human hypoxic AKI-on-a-Chip within a tight treatment window. Stem Cell Res Ther 2024; 15:105. [PMID: 38600585 PMCID: PMC11005291 DOI: 10.1186/s13287-024-03674-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/20/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Acute hypoxic proximal tubule (PT) injury and subsequent maladaptive repair present high mortality and increased risk of acute kidney injury (AKI) - chronic kidney disease (CKD) transition. Human bone marrow mesenchymal stem cell-derived exosomes (hBMMSC-Exos) as potential cell therapeutics can be translated into clinics if drawbacks on safety and efficacy are clarified. Here, we determined the real-time effective dose and treatment window of allogeneic hBMMSC-Exos, evaluated their performance on the structural and functional integrity of 3D microfluidic acute hypoxic PT injury platform. METHODS hBMMSC-Exos were isolated and characterized. Real-time impedance-based cell proliferation analysis (RTCA) determined the effective dose and treatment window for acute hypoxic PT injury. A 2-lane 3D gravity-driven microfluidic platform was set to mimic PT in vitro. ZO-1, acetylated α-tubulin immunolabelling, and permeability index assessed structural; cell proliferation by WST-1 measured functional integrity of PT. RESULTS hBMMSC-Exos induced PT proliferation with ED50 of 172,582 µg/ml at the 26th hour. Hypoxia significantly decreased ZO-1, increased permeability index, and decreased cell proliferation rate on 24-48 h in the microfluidic platform. hBMMSC-Exos reinforced polarity by a 1.72-fold increase in ZO-1, restored permeability by 20/45-fold against 20/155 kDa dextran and increased epithelial proliferation 3-fold compared to control. CONCLUSIONS The real-time potency assay and 3D gravity-driven microfluidic acute hypoxic PT injury platform precisely demonstrated the therapeutic performance window of allogeneic hBMMSC-Exos on ischemic AKI based on structural and functional cellular data. The novel standardized, non-invasive two-step system validates the cell-based personalized theragnostic tool in a real-time physiological microenvironment prior to safe and efficient clinical usage in nephrology.
Collapse
Affiliation(s)
- Sefa Burak Çam
- Faculty of Medicine, Dept. of Histology and Embryology, Hacettepe University, Ankara, Ankara, 06230, Turkey
| | - Eda Çiftci
- Graduate School of Science and Engineering, Department of Bioengineering, Hacettepe University, Ankara, 06230, Turkey
| | - Nazlıhan Gürbüz
- Graduate School of Science and Engineering, Department of Bioengineering, Hacettepe University, Ankara, 06230, Turkey
| | - Bülent Altun
- Faculty of Medicine, Dept. of Nephrology, Hacettepe University, Ankara, 06230, Turkey
| | - Petek Korkusuz
- Faculty of Medicine, Dept. of Histology and Embryology, Hacettepe University, Ankara, Ankara, 06230, Turkey.
| |
Collapse
|
5
|
Guimaraes APP, Calori IR, Stilhano RS, Tedesco AC. Renal proximal tubule-on-a-chip in PDMS: fabrication, functionalization, and RPTEC:HUVEC co-culture evaluation. Biofabrication 2024; 16:025024. [PMID: 38408383 DOI: 10.1088/1758-5090/ad2d2f] [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: 11/14/2023] [Accepted: 02/26/2024] [Indexed: 02/28/2024]
Abstract
'On-a-chip' technology advances the development of physiologically relevant organ-mimicking architecture by integrating human cells into three-dimensional microfluidic devices. This method also establishes discrete functional units, faciliting focused research on specific organ components. In this study, we detail the development and assessment of a convoluted renal proximal tubule-on-a-chip (PT-on-a-chip). This platform involves co-culturing Renal Proximal Tubule Epithelial Cells (RPTEC) and Human Umbilical Vein Endothelial Cells (HUVEC) within a polydimethylsiloxane microfluidic device, crafted through a combination of 3D printing and molding techniques. Our PT-on-a-chip significantly reduced high glucose level, exhibited albumin uptake, and simulated tubulopathy induced by amphotericin B. Remarkably, the RPTEC:HUVEC co-culture exhibited efficient cell adhesion within 30 min on microchannels functionalized with plasma, 3-aminopropyltriethoxysilane, and type-I collagen. This approach significantly reduced the required incubation time for medium perfusion. In comparison, alternative methods such as plasma and plasma plus polyvinyl alcohol were only effective in promoting cell attachment to flat surfaces. The PT-on-a-chip holds great promise as a valuable tool for assessing the nephrotoxic potential of new drug candidates, enhancing our understanding of drug interactions with co-cultured renal cells, and reducing the need for animal experimentation, promoting the safe and ethical development of new pharmaceuticals.
Collapse
Affiliation(s)
- Ana Paula Pereira Guimaraes
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Italo Rodrigo Calori
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Labs, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Oxford, MS 38677, United States of America
| | - Roberta Sessa Stilhano
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
| |
Collapse
|
6
|
Baker TK, Van Vleet TR, Mahalingaiah PK, Grandhi TSP, Evers R, Ekert J, Gosset JR, Chacko SA, Kopec AK. The Current Status and Use of Microphysiological Systems by the Pharmaceutical Industry: The International Consortium for Innovation and Quality Microphysiological Systems Affiliate Survey and Commentary. Drug Metab Dispos 2024; 52:198-209. [PMID: 38123948 DOI: 10.1124/dmd.123.001510] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Microphysiological systems (MPS) are comprised of one or multiple cell types of human or animal origins that mimic the biochemical/electrical/mechanical responses and blood-tissue barrier properties of the cells observed within a complex organ. The goal of incorporating these in vitro systems is to expedite and advance the drug discovery and development paradigm with improved predictive and translational capabilities. Considering the industry need for improved efficiency and the broad challenges of model qualification and acceptance, the International Consortium for Innovation and Quality (IQ) founded an IQ MPS working group in 2014 and Affiliate in 2018. This group connects thought leaders and end users, provides a forum for crosspharma collaboration, and engages with regulators to qualify translationally relevant MPS models. To understand how pharmaceutical companies are using MPS, the IQ MPS Affiliate conducted two surveys in 2019, survey 1, and 2021, survey 2, which differed slightly in the scope of definition of the complex in vitro models under question. The surveys captured demographics, resourcing, rank order for organs of interest, compound modalities tested, and MPS organ-specific questions, including nonclinical species needs and cell types. The major focus of this manuscript is on results from survey 2, where we specifically highlight the context of use for MPS within safety, pharmacology, or absorption, disposition, metabolism, and excretion and discuss considerations for including MPS data in regulatory submissions. In summary, these data provide valuable insights for developers, regulators, and pharma, offering a view into current industry practices and future considerations while highlighting key challenges impacting MPS adoption. SIGNIFICANCE STATEMENT: The application of microphysiological systems (MPS) represents a growing area of interest in the drug discovery and development framework. This study surveyed 20+ pharma companies to understand resourcing, current areas of application, and the key challenges and barriers to internal MPS adoption. These results will provide regulators, tech providers, and pharma industry leaders a starting point to assess the current state of MPS applications along with key learnings to effectively realize the potential of MPS as an emerging technology.
Collapse
Affiliation(s)
- Thomas K Baker
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.) baker_thomas_k@lilly
| | - Terry R Van Vleet
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Prathap Kumar Mahalingaiah
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Taraka Sai Pavan Grandhi
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Raymond Evers
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Jason Ekert
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - James R Gosset
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Silvi A Chacko
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Anna K Kopec
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| |
Collapse
|
7
|
Kimura H, Nakamura H, Goto T, Uchida W, Uozumi T, Nishizawa D, Shinha K, Sakagami J, Doi K. Standalone cell culture microfluidic device-based microphysiological system for automated cell observation and application in nephrotoxicity tests. LAB ON A CHIP 2024; 24:408-421. [PMID: 38131210 DOI: 10.1039/d3lc00934c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Microphysiological systems (MPS) offer an alternative method for culturing cells on microfluidic platforms to model organ functions in pharmaceutical and medical sciences. Although MPS hardware has been proposed to maintain physiological organ function through perfusion culture, no existing MPS can automatically assess cell morphology and conditions online to observe cellular dynamics in detail. Thus, with this study, we aimed to establish a practical strategy for automating cell observation and improving cell evaluation functions with low temporal resolution and throughput in MPS experiments. We developed a versatile standalone cell culture microfluidic device (SCCMD) that integrates microfluidic chips and their peripherals. This device is compliant with the ANSI/SLAS standards and has been seamlessly integrated into an existing automatic cell imaging system. This integration enables automatic cell observation with high temporal resolution in MPS experiments. Perfusion culture of human kidney proximal tubule epithelial cells using the SCCMD improves cell function. By combining the proximal tubule MPS with an existing cell imaging system, nephrotoxicity studies were successfully performed to automate morphological and material permeability evaluation. We believe that the concept of building the ANSI/SLAS-compliant-sized MPS device proposed herein and integrating it into an existing automatic cell imaging system for the online measurement of detailed cell dynamics information and improvement of throughput by automating observation operations is a novel potential research direction for MPS research.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Hiroko Nakamura
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Tomomi Goto
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Wakana Uchida
- Stem Cell Healthcare Business Unit, Nikon Corporation, Kanagawa, Japan
| | - Takayuki Uozumi
- Stem Cell Healthcare Business Unit, Nikon Corporation, Kanagawa, Japan
| | - Daniel Nishizawa
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Kenta Shinha
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Junko Sakagami
- Stem Cell Healthcare Business Unit, Nikon Corporation, Kanagawa, Japan
| | - Kotaro Doi
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan 153-8505
| |
Collapse
|
8
|
Brugger BA, Neuper L, Guettler J, Forstner D, Wernitznig S, Kummer D, Lyssy F, Feichtinger J, Krappinger J, El-Heliebi A, Bonstingl L, Moser G, Rodriguez-Blanco G, Bachkönig OA, Gottschalk B, Gruber M, Nonn O, Herse F, Verlohren S, Frank HG, Barapatre N, Kampfer C, Fluhr H, Desoye G, Gauster M. Fluid shear stress induces a shift from glycolytic to amino acid pathway in human trophoblasts. Cell Biosci 2023; 13:163. [PMID: 37684702 PMCID: PMC10492287 DOI: 10.1186/s13578-023-01114-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND The human placenta, a tissue with a lifespan limited to the period of pregnancy, is exposed to varying shear rates by maternal blood perfusion depending on the stage of development. In this study, we aimed to investigate the effects of fluidic shear stress on the human trophoblast transcriptome and metabolism. RESULTS Based on a trophoblast cell line cultured in a fluidic flow system, changes caused by shear stress were analyzed and compared to static conditions. RNA sequencing and bioinformatics analysis revealed an altered transcriptome and enriched gene ontology terms associated with amino acid and mitochondrial metabolism. A decreased GLUT1 expression and reduced glucose uptake, together with downregulated expression of key glycolytic rate-limiting enzymes, hexokinase 2 and phosphofructokinase 1 was observed. Altered mitochondrial ATP levels and mass spectrometry data, suggested a shift in energy production from glycolysis towards mitochondrial oxidative phosphorylation. This shift in energy production could be supported by increased expression of glutamic-oxaloacetic transaminase variants in response to shear stress as well as under low glucose availability or after silencing of GLUT1. The shift towards amino acid metabolic pathways could be supported by significantly altered amino acid levels, like glutamic acid, cysteine and serine. Downregulation of GLUT1 and glycolytic rate-limiting enzymes, with concomitant upregulation of glutamic-oxaloacetic transaminase 2 was confirmed in first trimester placental explants cultured under fluidic flow. In contrast, high fluid shear stress decreased glutamic-oxaloacetic transaminase 2 expression in term placental explants when compared to low flow rates. Placental tissue from pregnancies with intrauterine growth restriction are exposed to high shear rates and showed also decreased glutamic-oxaloacetic transaminase 2, while GLUT1 was unchanged and glycolytic rate-limiting enzymes showed a trend to be upregulated. The results were generated by using qPCR, immunoblots, quantification of immunofluorescent pictures, padlock probe hybridization, mass spectrometry and FRET-based measurement. CONCLUSION Our study suggests that onset of uteroplacental blood flow is accompanied by a shift from a predominant glycolytic- to an alternative amino acid converting metabolism in the villous trophoblast. Rheological changes with excessive fluidic shear stress at the placental surface, may disrupt this alternative amino acid pathway in the syncytiotrophoblast and could contribute to intrauterine growth restriction.
Collapse
Affiliation(s)
- Beatrice Anna Brugger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Jacqueline Guettler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Stefan Wernitznig
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Daniel Kummer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Freya Lyssy
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Julian Krappinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Lilli Bonstingl
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Giovanny Rodriguez-Blanco
- Clinical Institute for Medical and Chemical Laboratory Diagnosis, Medical University of Graz, Graz, Austria
| | - Olaf A Bachkönig
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Benjamin Gottschalk
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Michael Gruber
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Olivia Nonn
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Verlohren
- Department of Obstetrics and Gynaecology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Clinic for Obstetrics, Charité Berlin, Berlin, Germany
| | | | | | | | - Herbert Fluhr
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria.
| |
Collapse
|
9
|
Vilela de Sousa I, Ferreira MJS, Bebiano LB, Simões S, Matos AF, Pereira RF, Granja PL. Skin models of cutaneous toxicity, transdermal transport and wound repair. BURNS & TRAUMA 2023; 11:tkad014. [PMID: 37520659 PMCID: PMC10382248 DOI: 10.1093/burnst/tkad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/09/2023] [Accepted: 03/02/2023] [Indexed: 08/01/2023]
Abstract
Skin is widely used as a drug delivery route due to its easy access and the possibility of using relatively painless methods for the administration of bioactive molecules. However, the barrier properties of the skin, along with its multilayer structure, impose severe restrictions on drug transport and bioavailability. Thus, bioengineered models aimed at emulating the skin have been developed not only for optimizing the transdermal transport of different drugs and testing the safety and toxicity of substances but also for understanding the biological processes behind skin wounds. Even though in vivo research is often preferred to study biological processes involving the skin, in vitro and ex vivo strategies have been gaining increasing relevance in recent years. Indeed, there is a noticeably increasing adoption of in vitro and ex vivo methods by internationally accepted guidelines. Furthermore, microfluidic organ-on-a-chip devices are nowadays emerging as valuable tools for functional and behavioural skin emulation. Challenges in miniaturization, automation and reliability still need to be addressed in order to create skin models that can predict skin behaviour in a robust, high-throughput manner, while being compliant with regulatory issues, standards and guidelines. In this review, skin models for transdermal transport, wound repair and cutaneous toxicity will be discussed with a focus on high-throughput strategies. Novel microfluidic strategies driven by advancements in microfabrication technologies will also be revised as a way to improve the efficiency of existing models, both in terms of complexity and throughput.
Collapse
Affiliation(s)
| | | | - Luís B Bebiano
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal
- ISEP - Instituto Superior de Engenharia do Porto, Universidade do Porto, Rua Dr. António Bernardino de Almeida 431, 4200-072 Porto, Portugal
| | - Sandra Simões
- iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ana Filipa Matos
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rúben F Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | | |
Collapse
|
10
|
Wang Y, Gao Y, Pan Y, Zhou D, Liu Y, Yin Y, Yang J, Wang Y, Song Y. Emerging trends in organ-on-a-chip systems for drug screening. Acta Pharm Sin B 2023; 13:2483-2509. [PMID: 37425038 PMCID: PMC10326261 DOI: 10.1016/j.apsb.2023.02.006] [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: 11/18/2022] [Revised: 01/15/2023] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
New drug discovery is under growing pressure to satisfy the demand from a wide range of domains, especially from the pharmaceutical industry and healthcare services. Assessment of drug efficacy and safety prior to human clinical trials is a crucial part of drug development, which deserves greater emphasis to reduce the cost and time in drug discovery. Recent advances in microfabrication and tissue engineering have given rise to organ-on-a-chip, an in vitro model capable of recapitulating human organ functions in vivo and providing insight into disease pathophysiology, which offers a potential alternative to animal models for more efficient pre-clinical screening of drug candidates. In this review, we first give a snapshot of general considerations for organ-on-a-chip device design. Then, we comprehensively review the recent advances in organ-on-a-chip for drug screening. Finally, we summarize some key challenges of the progress in this field and discuss future prospects of organ-on-a-chip development. Overall, this review highlights the new avenue that organ-on-a-chip opens for drug development, therapeutic innovation, and precision medicine.
Collapse
Affiliation(s)
- Yanping Wang
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
- Sino-French Engineer School, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yanfeng Gao
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yongchun Pan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Dongtao Zhou
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yuta Liu
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yi Yin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yuzhen Wang
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Jiangsu National Synergistic Innovation Center for Advanced Materials, Nanjing Tech University, Nanjing 211816, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| |
Collapse
|
11
|
Priya PS, Guru A, Meenatchi R, Haridevamuthu B, Velayutham M, Seenivasan B, Pachaiappan R, Rajagopal R, Kuppusamy P, Juliet A, Arockiaraj J. Syringol, a wildfire residual methoxyphenol causes cytotoxicity and teratogenicity in zebrafish model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 864:160968. [PMID: 36549541 DOI: 10.1016/j.scitotenv.2022.160968] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Natural toxicants, particularly methoxy phenols (MPs) generated by wildfire lignin, can accumulate in the environment, and cause serious health hazards in living organisms. Although the toxicity of MPs such as guaiacol and catechol has recently been described, there is minimal evidence of ecotoxicological effects of syringol. As a result, this study focuses on determining the toxicity by evaluating the cytotoxic and teratogenic effects of syringol in vitro and in vivo in human embryonic kidney (HEK-293) cells and zebrafish embryos, respectively. The ecotoxicity of syringol was predicted to be 63.8 mg/L using the ECOSAR (ECOlogical Structure Activity Relationship) prediction tool, and molecular docking analysis was used to determine the interaction and binding affinities of syringol with human apoptotic proteins in silico. In HEK-293 cells, exposure of syringol (0.5-2 mg/L) has induced cytotoxicity in a concentration-dependent manner. In zebrafish larvae, exposure of syringol (0.5-2 mg/L) has induced dose-dependent embryo toxic effects (or growth abnormalities such as yolk sac edema, pericardial edema, skeletal abnormality, and hyperemia), and changes in growth morphometrics (head height, eye, yolk sac, and pericardial area, heart rate) in particular, the heart rate of larvae was found to be significantly decreased (p<0.001). After a 4-day experimental trial, the accumulated concentration of syringol in zebrafish larvae was confirmed both qualitatively (HPLC-MS - High Performance Liquid Chromatography-Mass spectrometry) and quantitatively (LC-QTOF-HRMS - Liquid Chromatography-Quadrupolar Time of Flight-High Resolution Mass spectrometry). The craniofacial abnormalities induced by syringol exposure (0.5-2 mg/L) were detected as anomalies in cartilaginous development and locomotor deficits using alcian blue staining and locomotor analyses, respectively. Significant increase in oxidative stress parameters (including reactive oxygen species generation, lipid peroxidation, superoxide dismutase, catalase, lactate dehydrogenase and nitric oxide production) (p<0.001) and substantial decrease in glutathione levels were observed (p<0.05) in syringol exposed zebrafish larvae through enzymatic analysis. Additionally, through acridine orange staining and gene expression analyses, syringol (2 mg/L) was found to activate apoptosis in zebrafish larvae. Considering the cytotoxic, embryotoxic (teratogenicity), and oxidative stress-related apoptotic effects of syringol in the zebrafish model, syringol has the potential to emerge as a potent environmental toxicant posing serious health hazards in many living systems; however, further research on its toxicological effects on the actual ecosystem and in higher animal models is required to confirm its consequences.
Collapse
Affiliation(s)
- P Snega Priya
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, 603 203 Chennai, Tamil Nadu, India
| | - Ajay Guru
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, SIMATS, 600 077 Chennai, Tamil Nadu, India
| | - Ramu Meenatchi
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, 603 203 Chennai, Tamil Nadu, India
| | - B Haridevamuthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, 603 203 Chennai, Tamil Nadu, India
| | - Manikandan Velayutham
- Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, SIMATS, 600 077, Chennai, Tamil Nadu, India
| | - Boopathi Seenivasan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, 603 203 Chennai, Tamil Nadu, India
| | - Raman Pachaiappan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Palaniselvam Kuppusamy
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, South Korea
| | - Annie Juliet
- Foundation for Aquaculture Innovations and Technology Transfer (FAITT), Thoraipakkam, Chennai 600 097, Tamil Nadu, India
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, 603 203 Chennai, Tamil Nadu, India.
| |
Collapse
|
12
|
Zhu Y, Shi Z, Ding W, Li C. On-chip construction of a fully structured scaffold-free vascularized renal tubule. Biomed Microdevices 2023; 25:8. [PMID: 36826720 DOI: 10.1007/s10544-023-00648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2023] [Indexed: 02/25/2023]
Abstract
Renal tubule chips have emerged as a promising platform for drug nephrotoxicity testing. However, the reported renal tubule chips hardly replicate the unique structure of renal tubules with thick proximal and distal tubules and a thin loop of Henle. In this study, we developed a fully structured scaffold-free vascularized renal tubule on a microfluidic chip. On the chip, the renal epithelial cell-laden hollow calcium-polymerized alginate tube with thick segments at both ends and a thin middle segment was U-shaped embedded in collagen hydrogel, parallel to the endothelial cell-laden hollow calcium-polymerized alginate tube with uniform tube diameter. After the alginate tubes were on-chip degraded, the renal epithelial cells and endothelial cells automatically attached to the collagen hydrogel and proliferated to form the renal tubule with proximal tubule, loop of Henle and distal tubule as well as peritubular blood vessel. We evaluated the viability of cells on the hollow alginate tubes, characterized the distribution and morphology of cells before and after the degradation of the alginate tube, and confirmed the proliferation of cells and the metabolic function of cells in terms of ATP synthesis, fibronectin secretion and VEGFR2 expression on the chip. The enhanced metabolic functions of renal epithelial cells and endothelial cells were preliminarily demonstrated. This study provides new insights into designing a more biomimetic renal tubule on a microfluidic chip.
Collapse
Affiliation(s)
- Yuntian Zhu
- , Hefei No.1 High School, 230041, Hefei, Anhui, China
| | - Zhengdi Shi
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Weiping Ding
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| | - Chengpan Li
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China. .,Center for Biomedical Imaging, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| |
Collapse
|
13
|
Zhang SY, Mahler GJ. A glomerulus and proximal tubule microphysiological system simulating renal filtration, reabsorption, secretion, and toxicity. LAB ON A CHIP 2023; 23:272-284. [PMID: 36514972 DOI: 10.1039/d2lc00887d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Microphysiological systems (MPS) are powerful predictive tools for assessing drug-induced kidney injuries. Previous MPS have examined single regions of the nephron, but lack simultaneous filtration, reabsorption, and secretion functionality. Here, we developed a partially open MPS that structurally and functionally recapitulated the glomerular filtration barrier, proximal tubular reabsorption, and secretion for seven days. The system introduced a recirculation circuit and an open filtrate output as a source of functional testing. As a proof-of-concept, a tri-culture of immortalized podocytes, umbilical vein endothelial cells, and proximal tubule (PCT) cells were housed in a single MPS: T-junction, glomerulus housing unit, and PCT chip. The MPS successfully retained blood serum protein, reabsorbed glucose, secreted creatinine, and expressed cell-type specific proteins (VE-cadherin, nephrin, and ZO-1). To simulate drug-induced kidney injuries, the system was perfused with cisplatin and adriamycin, and then tested using serum albumin filtration, glucose clearance, and lactate dehydrogenase release. The glomerulus and PCT MPS demonstrated a complex, dynamic microenvironment and recreated some in vivo-like functions in basal and drug-induced conditions, offering a novel prototype for preclinical testing.
Collapse
Affiliation(s)
- Stephanie Y Zhang
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY, 13902, USA.
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY, 13902, USA.
| |
Collapse
|
14
|
Lee JB, Kim H, Kim S, Sung GY. Fabrication and Evaluation of Tubule-on-a-Chip with RPTEC/HUVEC Co-Culture Using Injection-Molded Polycarbonate Chips. MICROMACHINES 2022; 13:mi13111932. [PMID: 36363953 PMCID: PMC9698344 DOI: 10.3390/mi13111932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 05/27/2023]
Abstract
To simulate the ADME process such as absorption, distribution, metabolism, and excretion in the human body after drug administration and to confirm the applicability of the mass production process, a microfluidic chip injection molded with polycarbonate (injection-molded chip (I-M chip)) was fabricated. Polycarbonate materials were selected to minimize drug absorption. As a first step to evaluate the I-M chip, RPTEC (Human Renal Proximal Tubule Epithelial Cells) and HUVEC (Human Umbilical Vein Endothelial Cells) were co-cultured, and live and dead staining, TEER (trans-epithelial electrical resistance), glucose reabsorption, and permeability were compared using different membrane pore sizes of 0.4 μm and 3 μm. Drug excretion was confirmed through a pharmacokinetic test with metformin and cimetidine, and the gene expression of drug transporters was confirmed. As a result, it was confirmed that the cell viability was higher in the 3 μm pore size than in the 0.4 μm, the cell culture performed better, and the drug secretion was enhanced when the pore size was large. The injection-molded polycarbonate microfluidic chip is anticipated to be commercially viable for drug screening devices, particularly ADME tests.
Collapse
Affiliation(s)
- Ju-Bi Lee
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Hyoungseob Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Sol Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
- Major in Materials Science and Engineering, School of Future Convergence, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
15
|
Parigoris E, Lee JH, Liu AY, Zhao X, Takayama S. Extended longevity geometrically-inverted proximal tubule organoids. Biomaterials 2022; 290:121828. [PMID: 36215909 PMCID: PMC10693433 DOI: 10.1016/j.biomaterials.2022.121828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/11/2022] [Accepted: 09/24/2022] [Indexed: 01/22/2023]
Abstract
This study reports the cellular self-organization of primary human renal proximal tubule epithelial cells (RPTECs) around a minimal Matrigel scaffold to produce basal-in and apical-out proximal tubule organoids (tubuloids). These tubuloids are produced and maintained in hanging drop cultures for 90+ days, the longest such culture of any kind reported to date. The tubuloids upregulate maturity markers, such as aquaporin-1 (AQP1) and megalin (LRP2), and exhibit less mesenchymal and proliferation markers, such as vimentin and Ki67, compared to 2D cultures. They also experience changes over time as revealed by a comparison of gene expression patterns of cells in 2D culture and in day 31 and day 67 tubuloids. Gene expression analysis and immunohistochemistry reveal an increase in the expression of megalin, an endocytic receptor that can directly bind and uptake protein or potentially assist protein uptake. The tubuloids, including day 90 tubuloids, uptake fluorescent albumin and reveal punctate fluorescent patterns, suggesting functional endocytic uptake through these receptors. Furthermore, the tubuloids release kidney injury molecule-1 (KIM-1), a common biomarker for kidney injury, when exposed to albumin in both dose- and time-dependent manners. While this study focuses on potential applications for modeling proteinuric kidney disease, the tubuloids may have broad utility for studies where apical proximal tubule cell access is required.
Collapse
Affiliation(s)
- Eric Parigoris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States; The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Ji-Hoon Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States; The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Amy Yunfan Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States; The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States; The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States.
| |
Collapse
|
16
|
Functional Evaluation and Nephrotoxicity Assessment of Human Renal Proximal Tubule Cells on a Chip. BIOSENSORS 2022; 12:bios12090718. [PMID: 36140103 PMCID: PMC9496563 DOI: 10.3390/bios12090718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022]
Abstract
An in vitro human renal proximal tubule model that represents the proper transporter expression and pronounced epithelial polarization is necessary for the accurate prediction of nephrotoxicity. Here, we constructed a high-throughput human renal proximal tubule model based on an integrated biomimetic array chip (iBAC). Primary human renal proximal tubule epithelial cells (hRPTECs) cultured on this microfluidic platform were able to form a tighter barrier, better transporter function and more sensitive nephrotoxicity prediction than those on the static Transwell. Compared with the human immortalized HK2 model, the hRPTECs model on the chip gained improved apical-basolateral polarization, barrier function and transporter expression. Polymyxin B could induce nephrotoxicity not only from the apical of the hRPTECs, but also from the basolateral side on the iBAC. However, other chemotherapeutic agents, such as doxorubicin and sunitinib, only induced nephrotoxicity from the apical surface of the hRPTECs on the iBAC. In summary, our renal proximal tubule model on the chip exhibits improved epithelial polarization and membrane transporter activity, and can be implemented as an effective nephrotoxicity-screening toolkit.
Collapse
|
17
|
Aceves JO, Heja S, Kobayashi K, Robinson SS, Miyoshi T, Matsumoto T, Schäffers OJM, Morizane R, Lewis JA. 3D proximal tubule-on-chip model derived from kidney organoids with improved drug uptake. Sci Rep 2022; 12:14997. [PMID: 36056134 PMCID: PMC9440090 DOI: 10.1038/s41598-022-19293-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/26/2022] [Indexed: 11/08/2022] Open
Abstract
Three-dimensional, organ-on-chip models that recapitulate kidney tissue are needed for drug screening and disease modeling. Here, we report a method for creating a perfusable 3D proximal tubule model composed of epithelial cells isolated from kidney organoids matured under static conditions. These organoid-derived proximal tubule epithelial cells (OPTECs) are seeded in cylindrical channels fully embedded within an extracellular matrix, where they form a confluent monolayer. A second perfusable channel is placed adjacent to each proximal tubule within these reusable multiplexed chips to mimic basolateral drug transport and uptake. Our 3D OPTEC-on-chip model exhibits significant upregulation of organic cation (OCT2) and organic anion (OAT1/3) transporters, which leads to improved drug uptake, compared to control chips based on immortalized proximal tubule epithelial cells. Hence, OPTEC tubules exhibit a higher normalized lactate dehydrogenase (LDH) release, when exposed to known nephrotoxins, cisplatin and aristolochic acid, which are diminished upon adding OCT2 and OAT1/3 transport inhibitors. Our integrated multifluidic platform paves the way for personalized kidney-on-chip models for drug screening and disease modeling.
Collapse
Affiliation(s)
- Jeffrey O Aceves
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Szilvia Heja
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kenichi Kobayashi
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Sanlin S Robinson
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Tomoya Miyoshi
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Takuya Matsumoto
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Olivier J M Schäffers
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Ryuji Morizane
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.
| | - Jennifer A Lewis
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
18
|
Kim H, Lee JB, Kim K, Sung GY. Effect of shear stress on the proximal tubule-on-a-chip for multi-organ microphysiological system. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
19
|
King J, Giselbrecht S, Truckenmüller R, Carlier A. Mechanistic Computational Models of Epithelial Cell Transporters-the Adorned Heroes of Pharmacokinetics. Front Pharmacol 2021; 12:780620. [PMID: 34803720 PMCID: PMC8599978 DOI: 10.3389/fphar.2021.780620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
Epithelial membrane transporter kinetics portray an irrefutable role in solute transport in and out of cells. Mechanistic models are used to investigate the transport of solutes at the organ, tissue, cell or membrane scale. Here, we review the recent advancements in using computational models to investigate epithelial transport kinetics on the cell membrane. Various methods have been employed to develop transport phenomena models of solute flux across the epithelial cell membrane. Interestingly, we noted that many models used lumped parameters, such as the Michaelis-Menten kinetics, to simplify the transporter-mediated reaction term. Unfortunately, this assumption neglects transporter numbers or the fact that transport across the membrane may be affected by external cues. In contrast, more recent mechanistic transporter kinetics models account for the transporter number. By creating models closer to reality researchers can investigate the downstream effects of physical or chemical disturbances on the system. Evidently, there is a need to increase the complexity of mechanistic models investigating the solute flux across a membrane to gain more knowledge of transporter-solute interactions by assigning individual parameter values to the transporter kinetics and capturing their dependence on each other. This change results in better pharmacokinetic predictions in larger scale platforms. More reliable and efficient model predictions can be made by creating mechanistic computational models coupled with dedicated in vitro experiments. It is also vital to foster collaborative efforts among transporter kinetics researchers in the modeling, material science and biological fields.
Collapse
Affiliation(s)
- Jasia King
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands.,Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
20
|
Kamat V, Robbings BM, Jung SR, Kelly J, Hurley JB, Bube KP, Sweet IR. Fluidics system for resolving concentration-dependent effects of dissolved gases on tissue metabolism. eLife 2021; 10:e66716. [PMID: 34734803 PMCID: PMC8660022 DOI: 10.7554/elife.66716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Oxygen (O2) and other dissolved gases such as the gasotransmitters H2S, CO, and NO affect cell metabolism and function. To evaluate effects of dissolved gases on processes in tissue, we developed a fluidics system that controls dissolved gases while simultaneously measuring parameters of electron transport, metabolism, and secretory function. We use pancreatic islets, retina, and liver from rodents to highlight its ability to assess effects of O2 and H2S. Protocols aimed at emulating hypoxia-reperfusion conditions resolved a previously unrecognized transient spike in O2 consumption rate (OCR) following replenishment of O2, and tissue-specific recovery of OCR following hypoxia. The system revealed both inhibitory and stimulatory effects of H2S on insulin secretion rate from isolated islets. The unique ability of this new system to quantify metabolic state and cell function in response to precise changes in dissolved gases provides a powerful platform for cell physiologists to study a wide range of disease states.
Collapse
Affiliation(s)
- Varun Kamat
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| | - Brian M Robbings
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Seung-Ryoung Jung
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| | | | - James B Hurley
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Kenneth P Bube
- Department of Mathematics, University of WashingtonSeattleUnited States
| | - Ian R Sweet
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| |
Collapse
|
21
|
Dadashzadeh A, Moghassemi S, Shavandi A, Amorim CA. A review on biomaterials for ovarian tissue engineering. Acta Biomater 2021; 135:48-63. [PMID: 34454083 DOI: 10.1016/j.actbio.2021.08.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Considerable challenges in engineering the female reproductive tissue are the follicle's unique architecture, the need to recapitulate the extracellular matrix, and tissue vascularization. Over the years, various strategies have been developed for preserving fertility in women diagnosed with cancer, such as embryo, oocyte, or ovarian tissue cryopreservation. While autotransplantation of cryopreserved ovarian tissue is a viable choice to restore fertility in prepubertal girls and women who need to begin chemo- or radiotherapy soon after the cancer diagnosis, it is not suitable for all patients due to the risk of having malignant cells present in the ovarian fragments in some types of cancer. Advances in tissue engineering such as 3D printing and ovary-on-a-chip technologies have the potential to be a translational strategy for precisely recapitulating normal tissue in terms of physical structure, vascularization, and molecular and cellular spatial distribution. This review first introduces the ovarian tissue structure, describes suitable properties of biomaterials for ovarian tissue engineering, and highlights recent advances in tissue engineering for developing an artificial ovary. STATEMENT OF SIGNIFICANCE: The increase of survival rates in young cancer patients has been accompanied by a rise in infertility/sterility in cancer survivors caused by the gonadotoxic effect of some chemotherapy regimens or radiotherapy. Such side-effect has a negative impact on these patients' quality of life as one of their main concerns is generating biologically related children. To aid female cancer patients, several research groups have been resorting to tissue engineering strategies to develop an artificial ovary. In this review, we discuss the numerous biomaterials cited in the literature that have been tested to encapsulate and in vitro culture or transplant isolated preantral follicles from human and different animal models. We also summarize the recent advances in tissue engineering that can potentially be optimal strategies for developing an artificial ovary.
Collapse
|
22
|
Viguerie A, Swapnasrita S, Veneziani A, Carlier A. A multi-domain shear-stress dependent diffusive model of cell transport-aided dialysis: analysis and simulation. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:8188-8200. [PMID: 34814295 DOI: 10.3934/mbe.2021406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Kidney dialysis is the most widespread treatment method for end-stage renal disease, a debilitating health condition common in industrialized societies. While ubiquitous, kidney dialysis suffers from an inability to remove larger toxins, resulting in a gradual buildup of these toxins in dialysis patients, ultimately leading to further health complications. To improve dialysis, hollow fibers incorporating a cell-monolayer with cultured kidney cells have been proposed; however, the design of such a fiber is nontrivial. In particular, the effects of fluid wall-shear stress have an important influence on the ability of the cell layer to transport toxins. In the present work, we introduce a model for cell-transport aided dialysis, incorporating the effects of the shear stress. We analyze the model mathematically and establish its well-posedness. We then present a series of numerical results, which suggest that a hollow-fiber design with a wavy profile may increase the efficiency of the dialysis treatment. We investigate numerically the shape of the wavy channel to maximize the toxin clearance. These results demonstrate the potential for the use of computational models in the study and advancement of renal therapies.
Collapse
Affiliation(s)
- Alex Viguerie
- Division of Mathematics, Gran Sasso Science Institute, Viale Francesco Crispi 7, L'Aquila, AQ 67100, Italy
| | - Sangita Swapnasrita
- Department of Cell-Biology Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Alessandro Veneziani
- Department of Mathematics, Emory University, 400 Dowman Drive, Atlanta, GA 30322, USA
- Department of Computer Science, Emory University, 400 Dowman Drive, Atlanta, GA 30322, USA
| | - Aurélie Carlier
- Department of Cell-Biology Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
23
|
Osório LA, Silva E, Mackay RE. A Review of Biomaterials and Scaffold Fabrication for Organ-on-a-Chip (OOAC) Systems. Bioengineering (Basel) 2021; 8:113. [PMID: 34436116 PMCID: PMC8389238 DOI: 10.3390/bioengineering8080113] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Drug and chemical development along with safety tests rely on the use of numerous clinical models. This is a lengthy process where animal testing is used as a standard for pre-clinical trials. However, these models often fail to represent human physiopathology. This may lead to poor correlation with results from later human clinical trials. Organ-on-a-Chip (OOAC) systems are engineered microfluidic systems, which recapitulate the physiochemical environment of a specific organ by emulating the perfusion and shear stress cellular tissue undergoes in vivo and could replace current animal models. The success of culturing cells and cell-derived tissues within these systems is dependent on the scaffold chosen; hence, scaffolds are critical for the success of OOACs in research. A literature review was conducted looking at current OOAC systems to assess the advantages and disadvantages of different materials and manufacturing techniques used for scaffold production; and the alternatives that could be tailored from the macro tissue engineering research field.
Collapse
Affiliation(s)
- Luana A. Osório
- Department of Mechanical, Aerospace and Civil Engineering, Brunel University London, Uxbridge UB8 3PH, UK;
| | - Elisabete Silva
- Department of Life Science, Brunel University London, Uxbridge UB8 3PH, UK;
| | - Ruth E. Mackay
- Department of Mechanical, Aerospace and Civil Engineering, Brunel University London, Uxbridge UB8 3PH, UK;
| |
Collapse
|
24
|
Three dimensional modeling of biologically relevant fluid shear stress in human renal tubule cells mimics in vivo transcriptional profiles. Sci Rep 2021; 11:14053. [PMID: 34234242 PMCID: PMC8263711 DOI: 10.1038/s41598-021-93570-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
The kidney proximal tubule is the primary site for solute reabsorption, secretion and where kidney diseases can originate, including drug-induced toxicity. Two-dimensional cell culture systems of the human proximal tubule cells (hPTCs) are often used to study these processes. However, these systems fail to model the interplay between filtrate flow, fluid shear stress (FSS), and functionality essential for understanding renal diseases and drug toxicity. The impact of FSS exposure on gene expression and effects of FSS at differing rates on gene expression in hPTCs has not been thoroughly investigated. Here, we performed RNA-sequencing of human RPTEC/TERT1 cells in a microfluidic chip-based 3D model to determine transcriptomic changes. We measured transcriptional changes following treatment of cells in this device at three different fluidic shear stress. We observed that FSS changes the expression of PTC-specific genes and impacted genes previously associated with renal diseases in genome-wide association studies (GWAS). At a physiological FSS level, we observed cell morphology, enhanced polarization, presence of cilia, and transport functions using albumin reabsorption via endocytosis and efflux transport. Here, we present a dynamic view of hPTCs response to FSS with increasing fluidic shear stress conditions and provide insight into hPTCs cellular function under biologically relevant conditions.
Collapse
|
25
|
Ren X, Getschman AE, Hwang S, Volkman BF, Klonisch T, Levin D, Zhao M, Santos S, Liu S, Cheng J, Lin F. Investigations on T cell transmigration in a human skin-on-chip (SoC) model. LAB ON A CHIP 2021; 21:1527-1539. [PMID: 33616124 PMCID: PMC8058301 DOI: 10.1039/d0lc01194k] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A microfluidics-based three-dimensional skin-on-chip (SoC) model is developed in this study to enable quantitative studies of transendothelial and transepithelial migration of human T lymphocytes in mimicked skin inflammatory microenvironments and to test new drug candidates. The keys results include 1) CCL20-dependent T cell transmigration is significantly inhibited by an engineered CCL20 locked dimer (CCL20LD), supporting the potential immunotherapeutic use of CCL20LD for treating skin diseases such as psoriasis; 2) transepithelial migration of T cells in response to a CXCL12 gradient mimicking T cell egress from the skin is significantly reduced by a sphingosine-1-phosphate (S1P) background, suggesting the role of S1P for T cell retention in inflamed skin tissues; and 3) T cell transmigration is induced by inflammatory cytokine stimulated epithelial cells in the SoC model. Collectively, the developed SoC model recreates a dynamic multi-cellular micro-environment that enables quantitative studies of T cell transmigration at a single cell level in response to physiological cutaneous inflammatory mediators and potential drugs.
Collapse
Affiliation(s)
- Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada. and Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Samuel Hwang
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - David Levin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Min Zhao
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA and Department of Ophthalmology & Vision Science, California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Susy Santos
- Victoria General Hospital, Winnipeg, MB R3T 2E8, Canada
| | - Song Liu
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Jasmine Cheng
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada.
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada. and Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada and Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA and Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
26
|
Kohl Y, Biehl M, Spring S, Hesler M, Ogourtsov V, Todorovic M, Owen J, Elje E, Kopecka K, Moriones OH, Bastús NG, Simon P, Dubaj T, Rundén-Pran E, Puntes V, William N, von Briesen H, Wagner S, Kapur N, Mariussen E, Nelson A, Gabelova A, Dusinska M, Velten T, Knoll T. Microfluidic In Vitro Platform for (Nano)Safety and (Nano)Drug Efficiency Screening. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006012. [PMID: 33458959 DOI: 10.1002/smll.202006012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Indexed: 06/12/2023]
Abstract
Microfluidic technology is a valuable tool for realizing more in vitro models capturing cellular and organ level responses for rapid and animal-free risk assessment of new chemicals and drugs. Microfluidic cell-based devices allow high-throughput screening and flexible automation while lowering costs and reagent consumption due to their miniaturization. There is a growing need for faster and animal-free approaches for drug development and safety assessment of chemicals (Registration, Evaluation, Authorisation and Restriction of Chemical Substances, REACH). The work presented describes a microfluidic platform for in vivo-like in vitro cell cultivation. It is equipped with a wafer-based silicon chip including integrated electrodes and a microcavity. A proof-of-concept using different relevant cell models shows its suitability for label-free assessment of cytotoxic effects. A miniaturized microscope within each module monitors cell morphology and proliferation. Electrodes integrated in the microfluidic channels allow the noninvasive monitoring of barrier integrity followed by a label-free assessment of cytotoxic effects. Each microfluidic cell cultivation module can be operated individually or be interconnected in a flexible way. The interconnection of the different modules aims at simulation of the whole-body exposure and response and can contribute to the replacement of animal testing in risk assessment studies in compliance with the 3Rs to replace, reduce, and refine animal experiments.
Collapse
Affiliation(s)
- Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Margit Biehl
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Sarah Spring
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Michelle Hesler
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Vladimir Ogourtsov
- Tyndall National Institute, University College Cork, Dyke Parade, Cork, T12 R5CP, Ireland
| | - Miomir Todorovic
- Tyndall National Institute, University College Cork, Dyke Parade, Cork, T12 R5CP, Ireland
| | - Joshua Owen
- Institute of Thermofluids, School of Mechanical Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | - Elisabeth Elje
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller, 2007, Norway
- Faculty of Medicine, Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Sognsvannsveien 9, Oslo, 0372, Norway
| | - Kristina Kopecka
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 84505, Slovakia
| | - Oscar Hernando Moriones
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and BIST, Campus UAB, Bellaterra 08193, Barcelona, Spain
- Universitat Autònoma de Barcelona (UAB), Campus UAB, Bellaterra, 08193, Barcelona, Spain
| | - Neus G Bastús
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and BIST, Campus UAB, Bellaterra 08193, Barcelona, Spain
| | - Peter Simon
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology SUT, Radlinskeho 9, Bratislava, 812 37, Slovakia
| | - Tibor Dubaj
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology SUT, Radlinskeho 9, Bratislava, 812 37, Slovakia
| | - Elise Rundén-Pran
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller, 2007, Norway
| | - Victor Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and BIST, Campus UAB, Bellaterra 08193, Barcelona, Spain
- Vall d'Hebron Institut de Recerca (VHIR), Barcelona, 08193, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08193, Spain
| | - Nicola William
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK
| | - Hagen von Briesen
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Sylvia Wagner
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Nikil Kapur
- Institute of Thermofluids, School of Mechanical Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | - Espen Mariussen
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller, 2007, Norway
| | - Andrew Nelson
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK
| | - Alena Gabelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 84505, Slovakia
| | - Maria Dusinska
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller, 2007, Norway
| | - Thomas Velten
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| | - Thorsten Knoll
- Fraunhofer Institute for Biomedical Engineering IBMT, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., Joseph-von-Fraunhofer-Weg 1, Sulzbach, 66280, Germany
| |
Collapse
|
27
|
Szymkowiak S, Sandler N, Kaplan DL. Aligned Silk Sponge Fabrication and Perfusion Culture for Scalable Proximal Tubule Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10768-10777. [PMID: 33621042 DOI: 10.1021/acsami.1c00548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chronic kidney disease and kidney failure are on the rise globally, yet there has not been a corresponding improvement in available therapies. A key challenge in a biological approach to developing kidney tissue is to identify scaffolding materials that support cell growth both in vitro and in vivo to facilitate translational goals. Scaffolds composed of silk fibroin protein possess the biocompatibility, mechanical robustness, and stability required for tissue engineering. Here, we use a silk sponge system to support kidney cells in a perfused bioreactor system. Silk fibroin protein underwent directional freezing to form parallel porous structures that mimic the native kidney structure of aligned tubules and are able to support more cells than nonaligned silk sponges. Adult immortalized renal proximal tubule epithelial cells were seeded into the sponges and cultured under static conditions for 1 week, then grown statically or with perfusion with culture media flowing through the sponge to enhance cell alignment and maturation. The sponges were imaged with confocal and scanning electron microscopies to analyze and quantify cell attachment, alignment, and expression of proteins important to proximal tubule differentiation and function. The perfused tissue constructs showed higher number of cells that are more evenly distributed through the construct and increased gene expression of several key markers of proximal tubule epithelial cell function compared to sponges grown under static conditions. These perfused tissue constructs represent a step toward a scalable approach to engineering proximal tubule structures with the potential to be used as in vitro models or as in vivo implantable tissues to supplement or replace impaired kidney function.
Collapse
Affiliation(s)
- Sophia Szymkowiak
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Nathan Sandler
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
28
|
Mansoorifar A, Gordon R, Bergan R, Bertassoni LE. Bone-on-a-chip: microfluidic technologies and microphysiologic models of bone tissue. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2006796. [PMID: 35422682 PMCID: PMC9007546 DOI: 10.1002/adfm.202006796] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 05/07/2023]
Abstract
Bone is an active organ that continuously undergoes an orchestrated process of remodeling throughout life. Bone tissue is uniquely capable of adapting to loading, hormonal, and other changes happening in the body, as well as repairing bone that becomes damaged to maintain tissue integrity. On the other hand, diseases such as osteoporosis and metastatic cancers disrupt normal bone homeostasis leading to compromised function. Historically, our ability to investigate processes related to either physiologic or diseased bone tissue has been limited by traditional models that fail to emulate the complexity of native bone. Organ-on-a-chip models are based on technological advances in tissue engineering and microfluidics, enabling the reproduction of key features specific to tissue microenvironments within a microfabricated device. Compared to conventional in-vitro and in-vivo bone models, microfluidic models, and especially organs-on-a-chip platforms, provide more biomimetic tissue culture conditions, with increased predictive power for clinical assays. In this review, we will report microfluidic and organ-on-a-chip technologies designed for understanding the biology of bone as well as bone-related diseases and treatments. Finally, we discuss the limitations of the current models and point toward future directions for microfluidics and organ-on-a-chip technologies in bone research.
Collapse
Affiliation(s)
- Amin Mansoorifar
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Ryan Gordon
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Luiz E. Bertassoni
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, USA
| |
Collapse
|
29
|
Troendle K, Rizzo L, Pichler R, Koch F, Itani A, Zengerle R, Lienkamp SS, Koltay P, Zimmermann S. Scalable fabrication of renal spheroids and nephron-like tubules by bioprinting and controlled self-assembly of epithelial cells. Biofabrication 2021; 13. [PMID: 33513594 DOI: 10.1088/1758-5090/abe185] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022]
Abstract
Scalable fabrication concepts of 3D kidney tissue models are required to enable their application in pharmaceutical high-throughput screenings. Yet the reconstruction of complex tissue structures remains technologically challenging. We present a novel concept reducing the fabrication demands, by using controlled cellular self-assembly to achieve higher tissue complexities from significantly simplified construct designs. We used drop-on-demand bioprinting to fabricate locally confined patterns of renal epithelial cells embedded in a hydrogel matrix. These patterns provide defined local cell densities (cell count variance < 11 %) with high viability (92 ± 2 %). Based on these patterns, controlled self-assembly leads to the formation of renal spheroids and nephron-like tubules with a predefined size and spatial localization. With this, we fabricated scalable arrays of hollow epithelial spheroids. The spheroid sizes correlated with the initial cell count per unit and could be stepwise adjusted, ranging from Ø = 84, 104, 120 to 131 µm in diameter (size variance < 9 %). Furthermore, we fabricated scalable line-shaped patterns, which self-assembled to hollow cellular tubules (Ø = 105 ± 22 µm). These showed a continuous lumen with prescribed orientation, lined by an epithelial monolayer with tight junctions. Additionally, upregulated expression of kidney-specific functional genes compared to 2D cell monolayers indicated increased tissue functionality, as revealed by mRNA sequencing. Furthermore, our concept enabled the fabrication of hybrid tubules, which consisted of arranged subsections of different cell types, combining murine and human epithelial cells. Finally, we integrated the self-assembled fabrication into a microfluidic chip and achieved fluidic access to the lumen at the terminal sites of the tubules. With this, we realized flow conditions with a wall shear stress of 0.05 ± 0.02 dyne/cm² driven by hydrostatic pressure for scalable dynamic culture towards a nephron-on-chip model.
Collapse
Affiliation(s)
- Kevin Troendle
- Department of Microsystems Engineering, Albert-Ludwigs-Universitat Freiburg, Fahnenbergplatz, Freiburg im Breisgau, 79085, GERMANY
| | - Ludovica Rizzo
- Institute of Anatomy and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Rämistrasse 71, Zurich, ZH, 8006, SWITZERLAND
| | - Roman Pichler
- Department of Nephrology, Universitätsklinikum Freiburg, Hugstetter Str. 55, Freiburg, 79106, GERMANY
| | - Fritz Koch
- Department of Microsystems Engineering, Albert-Ludwigs-Universitat Freiburg, Fahnenbergplatz, Freiburg im Breisgau, 79085, GERMANY
| | - Ahmad Itani
- Department of Microsystems Engineering, Albert-Ludwigs-Universitat Freiburg, Fahnenbergplatz, Freiburg im Breisgau, 79085, GERMANY
| | - Roland Zengerle
- Department of Microsystems Engineering, Albert-Ludwigs-Universitat Freiburg, Fahnenbergplatz, Freiburg im Breisgau, 79085, GERMANY
| | - Soeren S Lienkamp
- Institute of Anatomy and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Rämistrasse 71, Zurich, ZH, 8006, SWITZERLAND
| | - Peter Koltay
- Department of Microsystems Engineering, Albert-Ludwigs-Universitat Freiburg, Fahnenbergplatz, Freiburg im Breisgau, 79085, GERMANY
| | - Stefan Zimmermann
- Department of Microsystems Engineering, Albert-Ludwigs-Universitat Freiburg, Fahnenbergplatz, Freiburg im Breisgau, 79085, GERMANY
| |
Collapse
|
30
|
Mihevc M, Petreski T, Maver U, Bevc S. Renal proximal tubular epithelial cells: review of isolation, characterization, and culturing techniques. Mol Biol Rep 2020; 47:9865-9882. [PMID: 33170426 DOI: 10.1007/s11033-020-05977-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022]
Abstract
The kidney is a complex organ, comprised primarily of glomerular, tubular, mesangial, and endothelial cells, and podocytes. The fact that renal cells are terminally differentiated at 34 weeks of gestation is the main obstacle in regeneration and treatment of acute kidney injury or chronic kidney disease. Furthermore, the number of chronic kidney disease patients is ever increasing and with it the medical community should aim to improve existing and develop new methods of renal replacement therapy. On the other hand, as polypharmacy is on the rise, thought should be given into developing new ways of testing drug safety. A possible way to tackle these issues is with isolation and culture of renal cells. Several protocols are currently described to isolate the desired cells, of which the most isolated are the proximal tubular epithelial cells. They play a major role in water homeostasis, acid-base control, reabsorption of compounds, and secretion of xenobiotics and endogenous metabolites. When exposed to ischemic, toxic, septic, or obstructive conditions their death results in what we clinically perceive as acute kidney injury. Additionally, due to renal cells' limited regenerative potential, the profibrotic environment inevitably leads to chronic kidney disease. In this review we will focus on human proximal tubular epithelial cells. We will cover human kidney culture models, cell sources, isolation, culture, immortalization, and characterization subdivided into morphological, phenotypical, and functional characterization.
Collapse
Affiliation(s)
- Matic Mihevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
| | - Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Uroš Maver
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
| |
Collapse
|
31
|
Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintrón KM, McMinn P, Álvarez-García YR, Beebe DJ. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev 2020; 49:6402-6442. [PMID: 32760967 PMCID: PMC7521761 DOI: 10.1039/d0cs00705f] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microfluidic lumen-based systems are microscale models that recapitulate the anatomy and physiology of tubular organs. These technologies can mimic human pathophysiology and predict drug response, having profound implications for drug discovery and development. Herein, we review progress in the development of microfluidic lumen-based models from the 2000s to the present. The core of the review discusses models for mimicking blood vessels, the respiratory tract, the gastrointestinal tract, renal tubules, and liver sinusoids, and their application to modeling organ-specific diseases. We also highlight emerging application areas, such as the lymphatic system, and close the review discussing potential future directions.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - José M Ayuso
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Morgridge Institute for Research, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Yasmín R Álvarez-García
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
32
|
Shirazi J, Donzanti MJ, Nelson KM, Zurakowski R, Fromen CA, Gleghorn JP. Significant Unresolved Questions and Opportunities for Bioengineering in Understanding and Treating COVID-19 Disease Progression. Cell Mol Bioeng 2020; 13:259-284. [PMID: 32837585 PMCID: PMC7384395 DOI: 10.1007/s12195-020-00637-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
COVID-19 is a disease that manifests itself in a multitude of ways across a wide range of tissues. Many factors are involved, and though impressive strides have been made in studying this novel disease in a very short time, there is still a great deal that is unknown about how the virus functions. Clinical data has been crucial for providing information on COVID-19 progression and determining risk factors. However, the mechanisms leading to the multi-tissue pathology are yet to be fully established. Although insights from SARS-CoV-1 and MERS-CoV have been valuable, it is clear that SARS-CoV-2 is different and merits its own extensive studies. In this review, we highlight unresolved questions surrounding this virus including the temporal immune dynamics, infection of non-pulmonary tissue, early life exposure, and the role of circadian rhythms. Risk factors such as sex and exposure to pollutants are also explored followed by a discussion of ways in which bioengineering approaches can be employed to help understand COVID-19. The use of sophisticated in vitro models can be employed to interrogate intercellular interactions and also to tease apart effects of the virus itself from the resulting immune response. Additionally, spatiotemporal information can be gleaned from these models to learn more about the dynamics of the virus and COVID-19 progression. Application of advanced tissue and organ system models into COVID-19 research can result in more nuanced insight into the mechanisms underlying this condition and elucidate strategies to combat its effects.
Collapse
Affiliation(s)
- Jasmine Shirazi
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Michael J. Donzanti
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Katherine M. Nelson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Ryan Zurakowski
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| |
Collapse
|
33
|
Shinha K, Nihei W, Kimura H. A Microfluidic Probe Integrated Device for Spatiotemporal 3D Chemical Stimulation in Cells. MICROMACHINES 2020; 11:mi11070691. [PMID: 32708814 PMCID: PMC7408473 DOI: 10.3390/mi11070691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/05/2020] [Accepted: 07/14/2020] [Indexed: 11/16/2022]
Abstract
Numerous in vitro studies have been conducted in conventional static cell culture systems. However, most of the results represent an average response from a population of cells regardless of their local microenvironment. A microfluidic probe is a non-contact technology that has been widely used to perform local chemical stimulation within a restricted space, providing elaborated modulation and analysis of cellular responses within the microenvironment. Although microfluidic probes developed earlier have various potential applications, the two-dimensional structure can compromise their functionality and flexibility for practical use. In this study, we developed a three-dimensional microfluidic probe integrated device equipped with vertically oriented microchannels to overcome crucial challenges and tested the potential utility of the device in biological research. We demonstrated that the device tightly regulated spatial diffusion of a fluorescent molecule, and the flow profile predicted by simulation replicated the experimental results. Additionally, the device modulated the physiological Ca2+ response of cells within the restricted area by altering the local and temporal concentrations of biomolecules such as ATP. The novel device developed in this study may provide various applications for biological studies and contribute to further understanding of molecular mechanisms underlying cellular physiology.
Collapse
Affiliation(s)
- Kenta Shinha
- Department of Mechanical Engineering, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan; (K.S.); (W.N.)
| | - Wataru Nihei
- Department of Mechanical Engineering, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan; (K.S.); (W.N.)
- Micro/Nano Technology Center (MNTC), Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
| | - Hiroshi Kimura
- Department of Mechanical Engineering, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan; (K.S.); (W.N.)
- Micro/Nano Technology Center (MNTC), Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
- Correspondence: ; Tel.: +81-463-58-1211
| |
Collapse
|
34
|
Mazalan MB, Ramlan MAB, Shin JH, Ohashi T. Effect of Geometric Curvature on Collective Cell Migration in Tortuous Microchannel Devices. MICROMACHINES 2020; 11:E659. [PMID: 32630662 PMCID: PMC7408538 DOI: 10.3390/mi11070659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023]
Abstract
Collective cell migration is an essential phenomenon in many naturally occurring pathophysiological processes, as well as in tissue engineering applications. Cells in tissues and organs are known to sense chemical and mechanical signals from the microenvironment and collectively respond to these signals. For the last few decades, the effects of chemical signals such as growth factors and therapeutic agents on collective cell behaviors in the context of tissue engineering have been extensively studied, whereas those of the mechanical cues have only recently been investigated. The mechanical signals can be presented to the constituent cells in different forms, including topography, substrate stiffness, and geometrical constraint. With the recent advancement in microfabrication technology, researchers have gained the ability to manipulate the geometrical constraints by creating 3D structures to mimic the tissue microenvironment. In this study, we simulate the pore curvature as presented to the cells within 3D-engineered tissue-scaffolds by developing a device that features tortuous microchannels with geometric variations. We show that both cells at the front and rear respond to the varying radii of curvature and channel amplitude by altering the collective migratory behavior, including cell velocity, morphology, and turning angle. These findings provide insights into adaptive migration modes of collective cells to better understand the underlying mechanism of cell migration for optimization of the engineered tissue-scaffold design.
Collapse
Affiliation(s)
- Mazlee Bin Mazalan
- Graduate School of Engineering, Hokkaido University, Sapporo 060-8628, Japan;
- AMBIENCE, School of Microelectronic Engineering, Universiti Malaysia Perlis, Arau 02600, Perlis, Malaysia
| | | | - Jennifer Hyunjong Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science & Technology, Daejeon 34141, Korea;
| | - Toshiro Ohashi
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan;
| |
Collapse
|
35
|
Guenat OT, Geiser T, Berthiaume F. Clinically Relevant Tissue Scale Responses as New Readouts from Organs-on-a-Chip for Precision Medicine. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:111-133. [PMID: 31961712 DOI: 10.1146/annurev-anchem-061318-114919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Organs-on-chips (OOC) are widely seen as being the next generation in vitro models able to accurately recreate the biochemical-physical cues of the cellular microenvironment found in vivo. In addition, they make it possible to examine tissue-scale functional properties of multicellular systems dynamically and in a highly controlled manner. Here we summarize some of the most remarkable examples of OOC technology's ability to extract clinically relevant tissue-level information. The review is organized around the types of OOC outputs that can be measured from the cultured tissues and transferred to clinically meaningful information. First, the creation of functional tissues-on-chip is discussed, followed by the presentation of tissue-level readouts specific to OOC, such as morphological changes, vessel formation and function, tissue properties, and metabolic functions. In each case, the clinical relevance of the extracted information is highlighted.
Collapse
Affiliation(s)
- Olivier T Guenat
- ARTORG Center for Biomedical Engineering Research, Medical Faculty, University of Bern, CH-3008 Bern, Switzerland;
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
- Thoracic Surgery Department, University Hospital of Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
36
|
Phillips JA, Grandhi TSP, Davis M, Gautier JC, Hariparsad N, Keller D, Sura R, Van Vleet TR. A pharmaceutical industry perspective on microphysiological kidney systems for evaluation of safety for new therapies. LAB ON A CHIP 2020; 20:468-476. [PMID: 31989145 DOI: 10.1039/c9lc00925f] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The human kidney contains approximately one million nephrons. As the functional unit of the kidney, the nephron affords an opportunity to approximate the kidney at a microphysiological scale. Recent emergence of physiologically accurate human tissue models has radically advanced the possibilities of mimicking organ biology and multi-organ combinations in vitro. Anatomically, the nephron is one of the most complex, sequentially integrated microfluidic units in the body making the miniaturized microfluidic systems excellent candidates for capturing the kidney biology in vitro. While these models are promising, there are a number of considerations for practical implementation into a drug development paradigm. Opportunities for pharmaceutical industry applications of new MPS models often start with drug safety testing. As such, the intent of this article is to focus on safety and ADME applications. This article reviews biological functions of the kidney and options for characterizing known roles in nephrotoxicity. The concept of "context-of-use" is introduced as a framework for describing and verifying the specific features of an MPS platform for use in drug development. Overall, we present a perspective on key attributes of microphysiological kidney models, which the pharmaceutical industry could leverage to improve confident safety and ADME evaluations of experimental therapies.
Collapse
Affiliation(s)
| | - Taraka Sai Pavan Grandhi
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Myrtle Davis
- Bristol-Myers Squibb Company, Province Line Road, Princeton, New Jersey 08648, USA
| | | | | | - Douglas Keller
- Sanofi US, 55 Corporate Drive, Bridgewater, NJ 08807, USA
| | - Radhakrishna Sura
- Preclinical Safety, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA.
| | - Terry R Van Vleet
- Preclinical Safety, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA.
| |
Collapse
|
37
|
Three-dimensional cell-printing of advanced renal tubular tissue analogue. Biomaterials 2020; 232:119734. [DOI: 10.1016/j.biomaterials.2019.119734] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/13/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022]
|
38
|
Gottwald EM, Schuh CD, Drücker P, Haenni D, Pearson A, Ghazi S, Bugarski M, Polesel M, Duss M, Landau EM, Kaech A, Ziegler U, Lundby AKM, Lundby C, Dittrich PS, Hall AM. The iron chelator Deferasirox causes severe mitochondrial swelling without depolarization due to a specific effect on inner membrane permeability. Sci Rep 2020; 10:1577. [PMID: 32005861 PMCID: PMC6994599 DOI: 10.1038/s41598-020-58386-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
The iron chelator Deferasirox (DFX) causes severe toxicity in patients for reasons that were previously unexplained. Here, using the kidney as a clinically relevant in vivo model for toxicity together with a broad range of experimental techniques, including live cell imaging and in vitro biophysical models, we show that DFX causes partial uncoupling and dramatic swelling of mitochondria, but without depolarization or opening of the mitochondrial permeability transition pore. This effect is explained by an increase in inner mitochondrial membrane (IMM) permeability to protons, but not small molecules. The movement of water into mitochondria is prevented by altering intracellular osmotic gradients. Other clinically used iron chelators do not produce mitochondrial swelling. Thus, DFX causes organ toxicity due to an off-target effect on the IMM, which has major adverse consequences for mitochondrial volume regulation.
Collapse
Affiliation(s)
| | - Claus D Schuh
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Patrick Drücker
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Adam Pearson
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Susan Ghazi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - Michael Duss
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Ehud M Landau
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Anne K M Lundby
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Carsten Lundby
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland. .,Department of Nephrology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
39
|
Peired AJ, Mazzinghi B, De Chiara L, Guzzi F, Lasagni L, Romagnani P, Lazzeri E. Bioengineering strategies for nephrologists: kidney was not built in a day. Expert Opin Biol Ther 2020; 20:467-480. [DOI: 10.1080/14712598.2020.1709439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Benedetta Mazzinghi
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children’s University Hospital, Florence, Italy
| | - Letizia De Chiara
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Francesco Guzzi
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children’s University Hospital, Florence, Italy
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children’s University Hospital, Florence, Italy
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| |
Collapse
|
40
|
Dang BV, Taylor RA, Charlton AJ, Le-Clech P, Barber TJ. Toward Portable Artificial Kidneys: The Role of Advanced Microfluidics and Membrane Technologies in Implantable Systems. IEEE Rev Biomed Eng 2020; 13:261-279. [DOI: 10.1109/rbme.2019.2933339] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
41
|
Wan H, Gu C, Gan Y, Wei X, Zhu K, Hu N, Wang P. Sensor-free and Sensor-based Heart-on-a-chip Platform: A Review of Design and Applications. Curr Pharm Des 2019; 24:5375-5385. [PMID: 30734671 DOI: 10.2174/1381612825666190207170004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 01/09/2023]
Abstract
Drug efficacy and toxicity are key factors of drug development. Conventional 2D cell models or animal models have their limitations for the efficacy or toxicity assessment in preclinical assays, which induce the failure of candidate drugs or withdrawal of approved drugs. Human organs-on-chips (OOCs) emerged to present human-specific properties based on their 3D bioinspired structures and functions in the recent decade. In this review, the basic definition and superiority of OOCs will be introduced. Moreover, a specific OOC, heart-on-achip (HOC) will be focused. We introduce HOC modeling in the sensor-free and sensor-based way and illustrate the advantages of sensor-based HOC in detail by taking examples of recent studies. We provide a new perspective on the integration of HOC technology and biosensing to develop a new sensor-based HOC platform.
Collapse
Affiliation(s)
- Hao Wan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China.,State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Chenlei Gu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China.,State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Ying Gan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Xinwei Wei
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China.,State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Ning Hu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China.,State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China.,State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| |
Collapse
|
42
|
Szymkowiak S, Kaplan D. Biosynthetic Tubules: Multiscale Approaches to Kidney Engineering. CURRENT TRANSPLANTATION REPORTS 2019. [DOI: 10.1007/s40472-019-00248-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
43
|
Gottwald EM, Duss M, Bugarski M, Haenni D, Schuh CD, Landau EM, Hall AM. The targeted anti-oxidant MitoQ causes mitochondrial swelling and depolarization in kidney tissue. Physiol Rep 2019; 6:e13667. [PMID: 29611340 PMCID: PMC5880956 DOI: 10.14814/phy2.13667] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/07/2018] [Indexed: 12/30/2022] Open
Abstract
Kidney proximal tubules (PTs) contain a high density of mitochondria, which are required to generate ATP to power solute transport. Mitochondrial dysfunction is implicated in the pathogenesis of numerous kidney diseases. Damaged mitochondria are thought to produce excess reactive oxygen species (ROS), which can lead to oxidative stress and activation of cell death pathways. MitoQ is a mitochondrial targeted anti‐oxidant that has shown promise in preclinical models of renal diseases. However, recent studies in nonkidney cells have suggested that MitoQ might also have adverse effects. Here, using a live imaging approach, and both in vitro and ex vivo models, we show that MitoQ induces rapid swelling and depolarization of mitochondria in PT cells, but these effects were not observed with SS‐31, another targeted anti‐oxidant. MitoQ consists of a lipophilic cation (Tetraphenylphosphonium [TPP]) joined to an anti‐oxidant component (quinone) by a 10‐carbon alkyl chain, which is thought to insert into the inner mitochondrial membrane (IMM). We found that mitochondrial swelling and depolarization was also induced by dodecyltriphenylphosphomium (DTPP), which consists of TPP and the alkyl chain, but not by TPP alone. Surprisingly, MitoQ‐induced mitochondrial swelling occurred in the absence of a decrease in oxygen consumption rate. We also found that DTPP directly increased the permeability of artificial liposomes with a cardiolipin content similar to that of the IMM. In summary, MitoQ causes mitochondrial swelling and depolarization in PT cells by a mechanism unrelated to anti‐oxidant activity, most likely because of increased IMM permeability due to insertion of the alkyl chain.
Collapse
Affiliation(s)
| | - Michael Duss
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Claus D Schuh
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Ehud M Landau
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Musah S, Dimitrakakis N, Camacho DM, Church GM, Ingber DE. Directed differentiation of human induced pluripotent stem cells into mature kidney podocytes and establishment of a Glomerulus Chip. Nat Protoc 2019; 13:1662-1685. [PMID: 29995874 DOI: 10.1038/s41596-018-0007-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protocols have been established to direct the differentiation of human induced pluripotent stem (iPS) cells into nephron progenitor cells and organoids containing many types of kidney cells, but it has been difficult to direct the differentiation of iPS cells to form specific types of mature human kidney cells with high yield. Here, we describe a detailed protocol for the directed differentiation of human iPS cells into mature, post-mitotic kidney glomerular podocytes with high (>90%) efficiency within 26 d and under chemically defined conditions, without genetic manipulations or subpopulation selection. We also describe how these iPS cell-derived podocytes may be induced to form within a microfluidic organ-on-a-chip (Organ Chip) culture device to build a human kidney Glomerulus Chip that mimics the structure and function of the kidney glomerular capillary wall in vitro within 35 d (starting with undifferentiated iPS cells). The podocyte differentiation protocol requires skills for culturing iPS cells, and the development of a Glomerulus Chip requires some experience with building and operating microfluidic cell culture systems. This method could be useful for applications in nephrotoxicity screening, therapeutic development, and regenerative medicine, as well as mechanistic study of kidney development and disease.
Collapse
Affiliation(s)
- Samira Musah
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA. .,Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
45
|
Sjögren AK, Hornberg JJ. Compound selection and annotation to validate the predictivity of in vitro toxicity assays for use in drug discovery, in response to Commentary by Dr. Zink (Zink, D. Arch Toxicol (2018)). Arch Toxicol 2018; 93:225-226. [DOI: 10.1007/s00204-018-2359-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
|
46
|
Gabelova A, Kozics K, Kapka-Skrzypczak L, Kruszewski M, Sramkova M. Nephrotoxicity: Topical issue. Mutat Res 2018; 845:402988. [PMID: 31561894 DOI: 10.1016/j.mrgentox.2018.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022]
Abstract
Drug-induced kidney injury is one of the most significant adverse events and dose limiting factor in chemotherapy as well a major cause of prospective drug attrition during pharmaceutical development. Moreover, kidney injury can also occur as a consequence of exposures to environmental xenobiotics such as heavy metals, fungal toxins and nanomaterials. The lack of adequate in vitro human kidney models that mimic more realistically the in vivo conditions and the absence of suitable and robust, cost-effective and predictive cell-based in vitro assays contribute to an underestimation of the kidney toxic potential of new drugs and xenobiotics. Therefore, a rapid screening system capable to detect potential nephrotoxicity at early stages of drug discovery is an urgent need. Here we provide an overview of human cell lines currently used as a surrogate in vitro kidney models in nephrotoxicity studies, including their advantages and limitations. In addition, the capacity of the single cell gel electrophoresis (SCGE)/comet assay as a potential tool in kidney toxicants screening is discussed. Despite a limited number of studies using the comet assay to evaluate the drug-induced kidney damage potential, a considerable variability in SCGE methodology (e.g. lysis, unwinding, and electrophoresis conditions) has been observed. Before the comet assay can be included in nephrotoxicity testing, a basic guideline has to be developed. To test its feasibility, additional in vitro experiments including inter-laboratory validation studies based on this guideline have to be performed.
Collapse
Affiliation(s)
- Alena Gabelova
- Cancer Research Institute, Biomedical Research Center SAS, Dubravska cesta 9, 845 05 Bratislava, Slovakia.
| | - Katarina Kozics
- Cancer Research Institute, Biomedical Research Center SAS, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Lucyna Kapka-Skrzypczak
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, Sucharskiego 2, 35-225, Rzeszów, Poland
| | - Marcin Kruszewski
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, Sucharskiego 2, 35-225, Rzeszów, Poland; Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195, Warsaw, Poland
| | - Monika Sramkova
- Cancer Research Institute, Biomedical Research Center SAS, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
47
|
Rayner SG, Phong KT, Xue J, Lih D, Shankland SJ, Kelly EJ, Himmelfarb J, Zheng Y. Reconstructing the Human Renal Vascular-Tubular Unit In Vitro. Adv Healthc Mater 2018; 7:e1801120. [PMID: 30379416 PMCID: PMC6478624 DOI: 10.1002/adhm.201801120] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Indexed: 12/19/2022]
Abstract
Engineered human kidney-on-a-chip platforms show tremendous promise for disease modeling and drug screening. Outstanding challenges exist, however, in reconstructing the complex architecture, cellular make-up, and matrix composition necessary for the proper modeling of kidney function. Herein, the first fully tunable human kidney-on-a-chip platform is reported that allows the reconstruction of the native architecture of the renal endothelial-epithelial exchange interface using entirely cell-remodelable matrix and patient-derived kidney cells. This platform consists of a double-layer human renal vascular-tubular unit (hRVTU) enabled by a thin collagen membrane that replicates the kidney exchange interface. It is shown that endothelial and epithelial cells lining their respective lumens remodel the membrane in culture into a ≈1 µm thick exchange interface composed of native basement membrane proteins. This interface displays sufficient mechanical integrity for media flow and blood perfusion. As a proof of principle, it is demonstrated that the hRVTU performs kidney-specific functions including reabsorption of albumin and glucose from the epithelial channel. By incorporating multiple cell populations from single donors, it is demonstrated that the hRVTU may have utility for future precision medicine applications. The success of the system provides new opportunities for the next generation of organ-on-a-chip models.
Collapse
Affiliation(s)
- Samuel G. Rayner
- Department of Bioengineering, University of Washington, Seattle, Washington 98109
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington 98109
| | - Kiet T Phong
- Department of Bioengineering, University of Washington, Seattle, Washington 98109
| | - Jun Xue
- Department of Bioengineering, University of Washington, Seattle, Washington 98109
| | - Daniel Lih
- Department of Bioengineering, University of Washington, Seattle, Washington 98109
| | - Stuart J. Shankland
- Department of Medicine, University of Washington, Seattle, Washington 98109
- Kidney Research Institute, University of Washington, Seattle, Washington 98109
| | - Edward J. Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98109
- Kidney Research Institute, University of Washington, Seattle, Washington 98109
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109
| | - Jonathan Himmelfarb
- Department of Bioengineering, University of Washington, Seattle, Washington 98109
- Department of Medicine, University of Washington, Seattle, Washington 98109
- Kidney Research Institute, University of Washington, Seattle, Washington 98109
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, Washington 98109
- Kidney Research Institute, University of Washington, Seattle, Washington 98109
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109
| |
Collapse
|
48
|
|
49
|
Mittal R, Woo FW, Castro CS, Cohen MA, Karanxha J, Mittal J, Chhibber T, Jhaveri VM. Organ‐on‐chip models: Implications in drug discovery and clinical applications. J Cell Physiol 2018; 234:8352-8380. [DOI: 10.1002/jcp.27729] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Frank W. Woo
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Carlo S. Castro
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Madeline A. Cohen
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Joana Karanxha
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Jeenu Mittal
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Tanya Chhibber
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University Chandigarh India
| | - Vasanti M. Jhaveri
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
50
|
Caetano-Pinto P, Stahl SH. Perspective on the Application of Microphysiological Systems to Drug Transporter Studies. Drug Metab Dispos 2018; 46:1647-1657. [DOI: 10.1124/dmd.118.082750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022] Open
|